ENTERIC GLIAL MODULATION OF IMMUNE ACTIVATION DURING INFLAMMATORY STRESS By Aaron Kin Yeung Chow A DISSERTATION Michigan State University in partial fulfillment of the requirements Submitted to for the degree of Physiology–Doctor of Philosophy 2020 ABSTRACT ENTERIC GLIAL MODULATION OF IMMUNE ACTIVATION DURING INFLAMMATORY STRESS By Aaron Kin Yeung Chow Gastrointestinal (GI) disorders such as inflammatory bowel disease (IBD), irritable bowel syndrome, and other functional GI disorders are major health concerns and account for about $26.4 billion in yearly costs in the United States alone. Many of these GI disorders manifest symptoms such as GI dysmotility, intestinal secretion and absorption dysfunction, and abdominal pain. Inflammation plays a major role in the pathogenesis of these diseases, and current therapies for many GI disorders aim to regulate the activation and progression of the inflammatory cascade. However, due to a gap in knowledge in how the immune system is regulated within the GI tract, there remains a lack of effective treatments for these common GI disorders. Interactions between the nervous system and immune system point to neurons having important roles in immune modulation, but the mechanisms of neuro-immune regulation in the gut is not completely understood. The enteric nervous system (ENS) consists of enteric neurons and enteric glia arranged in plexuses embedded in the gut wall. This neural network is responsible for the normal secretomotor functioning of the GI tract, and the disruption of the ENS network alters GI functioning and underlies pathological GI symptoms. As part of the ENS, enteric glia work in tandem with enteric neurons to coordinate GI functions. In addition to their contributions to maintain normal secretomotor functioning of the GI tract, enteric glia are activated by immunomodulatory signals, they can secrete and respond to cytokines, can exert immunosuppressive effects, and share characteristics with antigen presenting cells. Therefore, we hypothesize that enteric glia play an active role in immune regulation in the ENS. In this dissertation, we specifically examine the role enteric glial cells play as an antigen presenting cell to regulate immune activation. Our results show that enteric glia have the machinery necessary to act as an antigen presenting cell and can express major histocompatibility complex (MHC) type II molecules during inflammatory stress to interact with T-lymphocytes. Enteric glial MHC II expression has functional relevance, as it modulates the activation in Th17 and Treg subtypes, but not Th1 or Th2 T- lymphocyte subtypes. Although MHC II molecules are typically associated with the expression of phagocytosed extracellular antigens, our results show that enteric glia do not readily phagocytose extracellular antigens. Instead, MHC II expression in enteric glia is mediated by autophagy. The activation of autophagic pathways is necessary, but not sufficient in eliciting enteric glial MHC II expression. Finally, although enteric glia regulate T-lymphocyte activation, cytokine levels at the whole organism or regional tissue levels remain unchanged, suggesting that enteric glial cytokine effects primarily operate at the local microenvironment level. Our findings provide support for enteric glial cells having an active role as an immunomodulator. Specifically, we show that enteric glia modulate T-lymphocyte activation via autophagy-mediated MHC II expression and propose a novel mechanism of neuro-immune modulation in the gut. ACKNOWLEDGEMENTS I would like to first thank my mentor Dr. Brian Gulbransen for the guidance he has given me in this journey. His support and mentorship on how to become an efficient and successful scientist has brought me to where I am today in my research career. His positive outlook on any given situation has encouraged me to never give up even when faced with discouraging experimental results. Brian always had time to sit down to discuss any concerns I had whether it was science-related or otherwise, and he was never too busy to talk about data. I am especially grateful for the patience and understanding Brian has for the DO-PhD program – the sheer length of the program, the extra obligations I have that pull me away from experiments, and the times during medical school where I spent studying and wasn’t very productive in lab. I would also like to thank the past and present members of the Gulbransen Lab who have made being part of the lab such an enjoyable experience. Jonathan, David, Vlado, Siomara, Isola, Ninotchska, Wilmarie, Luisa, Christine, Mustafa, RJ, Becca, and Erika, thank you for all the help in lab and experiments when I overestimate how much I can manage in one day. Thank you for your insights and and helping me develop as a scientist, and our time spent hanging out outside of lab as well. Your help and friendship have made my experiences here very enjoyable. I would also like to thank the DO-PhD program who made my graduate career here at MSU possible. Thank you, Dr. Justin McCormick and Ms. Bethany Heinlen, for admitting me to the DO-PhD program and allowing me on this wonderful journey. Thank you to Dr. Brian Schutte and Ms. Michelle Volker who are the current DO-PhD iv administrators who continue to run the DO-PhD program and who are supportive of my journey. I would also like to thank the BMS program, especially Becky and Jasmine, who have been extremely patient and helpful as I navigate the intricacies of graduate school. It is true that time flies when you’re having fun, and I would like to thank the friends that I have made along the way here in Michigan that have made it feel like I moved to Michigan only a few short weeks ago. I would like to especially thank Briana, Laura, Sean, Scott, and Yajing – the times that we have shared together and friendship that we have makes this long journey so much fun. I would like to thank my friends and family back in Canada as well, who have been ever supportive of my studies and studies here in the United States, no matter the physical distance that separate us, or the sheer number of years that I have spent as a student. They have never ceased supporting my endeavors here and have been my biggest supporters, reminding me during stressful times that it will all be worth it in the end. I would also like to thank my committee members Dr. Julia Busik, Dr. James Galligan, Dr. Linda Mansfield, Dr. Adam Moeser, and Dr. Narayanan Parameswaran who have helped me in refining my project, giving me valuable feedback, and your attention during the countless hours of committee meetings. Your support has helped guide my development as a scientist. v I could not have been able to accomplish this journey without the help and support of family, friends, staff, faculty, collaborators, and countless others that are not mentioned here. Thank you from the bottom of my heart. vi TABLE OF CONTENTS LIST OF TABLES ............................................................................................................ x LIST OF FIGURES ..........................................................................................................xi KEY TO ABBREVIATIONS ........................................................................................... xiii CHAPTER 1: Introduction ............................................................................................... 1 Overview .............................................................................................................. 2 The Enteric Nervous System ................................................................................ 3 Intrinsic ENS innervation ............................................................................ 3 Extrinsic inputs into the ENS ...................................................................... 7 ENS damage and disease ......................................................................... 9 Enteric Glia .............................................................................................. 13 The Immune System .......................................................................................... 17 Inflammation ............................................................................................ 19 The Innate Immune System ..................................................................... 22 Antigen Presenting Cells .......................................................................... 24 Type II Major Histocompatibility Complex ................................................ 26 The Adaptive Immune System ................................................................. 31 T-lymphocytes ......................................................................................... 33 Current models of intestinal inflammation ................................................ 35 Summary and Aims of Dissertation .......................................................... 37 REFERENCES ................................................................................................... 39 CHAPTER 2: Potential roles of enteric glia in bridging neuroimmune communication in the gut ........................................................................................................................... 58 Abstract .............................................................................................................. 59 Introduction ......................................................................................................... 60 Contribution of enteric neurons to inflammation ....................................... 61 Glia in the healthy intestine ...................................................................... 66 Glial cells are activated by cytokines and immunomodulatory signals ..... 68 Glia express and secrete cytokines and pro-inflammatory signals .......... 72 Glial cells act as antigen presenting cells ................................................ 74 Enteric glia exert immunosuppressive and anti-inflammatory effects ...... 75 Conclusions ........................................................................................................ 77 REFERENCES ................................................................................................... 79 vii CHAPTER 3: Enteric glial cells induce Th17 and Treg activation via autophagy-mediated MHC II expression ......................................................................................................... 86 Abstract .............................................................................................................. 87 Background and Aims .............................................................................. 87 Methods ................................................................................................... 87 Results ..................................................................................................... 87 Conclusions ............................................................................................. 88 Introduction ......................................................................................................... 89 Materials and Methods ....................................................................................... 91 Animals .................................................................................................... 91 Induction of glial MHC II ........................................................................... 92 Phagocytosis assay ................................................................................. 92 Whole-Mount Immunofluorescence ......................................................... 93 H&E Staining ........................................................................................... 94 Frozen section Immunofluorescence ....................................................... 94 Flow cytometry ......................................................................................... 95 Gene expression ...................................................................................... 96 Cytokine quantification ............................................................................. 98 Autophagy induction and detection .......................................................... 99 Data Analysis ........................................................................................... 99 Statistics ................................................................................................ 101 Results ............................................................................................................. 104 Conditional ablation of glial MHC II in Sox10CreERT2;IABfl/fl mice and IFNγ + LPS challenge ........................................................................................ 104 Enteric glia express mature MHC II in response to IFNγ + LPS ............ 109 Enteric glial MHC II expression contributes to CD4+ T-cell activation in mesenteric lymph nodes ........................................................................ 112 Enteric glia affect local, but not serum or colonic cytokine levels ........... 120 Pro-inflammatory stimuli IFNγ and LPS do not trigger phagocytosis in enteric glia ............................................................................................. 132 Glial autophagy regulates MHC II expression ........................................ 134 Discussion ........................................................................................................ 139 REFERENCES ................................................................................................. 147 CHAPTER 4: Summary, Key Findings and Future Directions ..................................... 157 Summary .......................................................................................................... 158 Key Findings ..................................................................................................... 158 Enteric glial cells express MHC II in response to low dose IFNγ and LPS ............................................................................................................... 158 Enteric glial MHC II alter T-lymphocyte activation .................................. 159 Enteric glial MHC II expression is derived from autophagy pathways .... 160 Study Limitations .............................................................................................. 161 Pharmacological modulators .................................................................. 161 viii Sex of experimental animals .................................................................. 161 MHC II knockout animals ....................................................................... 161 Future Directions .............................................................................................. 161 Determination of antigens presented on enteric glial MHC II ................. 161 Enteric glial autophagy effects in modulating T-lymphocyte activation .. 162 Glial MHC II contributions in overt inflammation .................................... 162 Sympathetic effects on enteric glial-mediated T-lymphocyte activation . 163 Local renin angiotensin effects .............................................................. 165 REFERENCES ................................................................................................. 169 ix LIST OF TABLES Table 3.1: Antibodies used in Chapter 3 ..................................................................... 102 x LIST OF FIGURES Figure 1.1: Arrangement of the ENS within the GI tract .................................................. 6 Figure 1.2: Enteric glial proximity to enteric neurons ..................................................... 14 Figure 1.3: Overview of enteric glial functions ............................................................... 16 Figure 1.4: Overview of typical GI immunity .................................................................. 18 Figure 1.5: Murine MHC II. ............................................................................................ 27 Figure 1.6: Maturation of MHC II molecules .................................................................. 29 Figure 2.1: Overview of enteric neuron responses to select signals ............................. 64 Figure 2.2: Overview of enteric glial cell responses to select signals ............................ 70 Figure 3.1: Enteric glial cells express MHC II when stimulated with IFNγ and LPS .... 105 Figure 3.2: Exposure to IFNγ and LPS causes weight loss but does not cause significant tissue damage or observable inflammation ................................................ 107 Figure 3.3: Pattern and level of MHC II expression in macrophages does not change when treated with IFNγ and LPS ................................................................................. 111 Figure 3.4: CD4 expressing T-lymphocytes are in close proximity with GFAP expressing enteric glia in the myenteric plexus ............................................................................. 115 Figure 3.5: Enteric glial MHC II expression affects T-lymphocyte activation ............... 116 Figure 3.6: Activation of CD8+ T-lymphocytes decreased when MHC II ablated in enteric glia ................................................................................................................... 118 Figure 3.7: Enteric glial MHC II expression affect FoxP3+ and IL-17+ cell populations to a greater degree than IFNγ+ and IL-4+ cell populations ............................................. 119 Figure 3.8: Enteric glial gene expression .................................................................... 122 Figure 3.9: Enteric glial MHC II expression has limited effects on serum and colonic tissue cytokine levels ................................................................................................... 124 Figure 3.10: MHC II ablation in enteric glial cells do not show changes in colonic tissue level of select cytokines ............................................................................................... 126 xi Figure 3.11: MHC II ablation in enteric glial cells do not show changes in serum level of select cytokines ........................................................................................................... 128 Figure 3.12: Sox10 expressing cells are present in sparse numbers in spleen tissue and express MHC II ........................................................................................................... 130 Figure 3.13: Enteric glia do not exhibit phagocytic activity when stimulated with IFNγ and LPS ...................................................................................................................... 133 Figure 3.14: Enteric glia express MHC II via autophagy pathways ............................. 136 Figure 3.15: Schematic mechanism of enteric glial MHC II expression ....................... 146 Figure 4.1: Autonomic innervation onto enteric glia may modulate MHC II expression .................................................................................................................................... 164 Figure 4.2: The renin-angiotensin pathway neuronal damage and inflammation ........ 167 Figure 4.3: Enteric glia increase expression of ACE2 during high fat diet conditions .. 168 xii KEY TO ABBREVIATIONS 3-MA – 3-methyladenine 6-OHDA – 6-hydroxydopamine CD – Cluster of differentiation CNS – Central nervous system DMEM – Dulbecco’s Modified Eagle Medium EDNRB – Endothelin B receptor ENS – Enteric nervous system FACS – Fluorescence-activated cell sorting GDNF – Glial derived neurotrophic factor GFAP – Glial fibrillary acidic protein GI – Gastrointestinal GSNO – Glial derived S-nitrosoglutatione IBD – Inflammatory bowel disease IBS – Irritable bowel syndrome IFN-γ – Interferon gamma IL – Interleukin IP – Intraperitoneal xiii KO – Knockout LMMP – Longitudinal muscle-myenteric plexus LPS – Lipopolysaccharide MAPK – Mitogen-activated protein kinase MHC – Major histocompatibility complex NF-κB – Nuclear factor kappa-light-chain-enhancer of activated B cells PBS – Phosphate buffered saline TGF-β – Transforming growth factor beta TLR – Toll-like receptor TNBS – Trinitrobenzene sulfonic acid TNF – Tumor necrosis factor VIP – Vasoactive intestinal peptide WT – Wildtype xiv CHAPTER 1: Introduction 1 Overview The gastrointestinal (GI) tract is an important organ system responsible for the movement, digestion, absorption, excretion, and barrier defense against ingested particles (60, 128). The proper functioning of the GI tract is coordinated primarily by the enteric nervous system (ENS) (60), with modulating inputs from the central nervous system (CNS) (60). The ENS is a part of the autonomic nervous system and is embedded within the walls of the GI tract, and dysfunction in the ENS is a large component to the development of varying GI disorders (57). The loss of neuronal inputs into the GI tract leads to uncoordinated GI function, dysmotility, improper GI secretion, absorption, and improper regulation in GI blood flow (56). ENS neuropathies can be due to congenital causes, effects from the surrounding environment or disease, or a combination of factors (56, 57), leading pathological GI findings in diseases such as in Hirschsprung’s disease, Parkinson’s disease, Inflammatory bowel disease (IBD), Diabetes mellitus, Chronic intestinal pseudo-obstruction, Chagas disease, among others (56, 57, 148, 202). Inflammation plays a major role in neuronal damage and death in the ENS, and is the hallmark finding in IBD. Given that the lumen of the GI tract is continuous with the outside environment, the constant exposure to bacteria, viruses, parasites, and other foreign particles are major sources for potential inflammatory stimuli (60, 192). Therefore, the GI tract plays a critical role in pathogen defense and is a major site for immune regulation (60, 143, 149). Although components such as the microbiome has been the focus of many studies examining GI immune regulation, there is growing support that the ENS has a major role in the modulating immune activity (114, 219). 2 Increasing evidence show that both enteric neurons and enteric glia, which are non- myelinating peripheral glia in the ENS, play substantial roles in immune regulation as well (22, 114). Specifically, enteric glia have the prerequisite cellular machinery to directly interact with the immune system as an antigen presenting cell and modulate its activity (4, 53, 90, 201). However, many of the studies have been performed in vitro, or have not shown functional effects on the immune system. Therefore, the specific roles that enteric glia play in immune regulation is not well understood. Here, this dissertation examines enteric glial function and responses in inflammation and its role in immune modulation in the ENS. Intrinsic ENS innervation The Enteric Nervous System The ENS, also referred to the “brain of the gut”, is part of the autonomic nervous system (48) and is embedded along the entire length of the GI tract. The ENS receives inputs from the CNS but is functionally independent (48, 97). In fact, the GI tract maintains coordinated activity and does not cease to function when completely separated from the CNS (6, 7). The ENS is composed primarily of two ganglionated plexuses, termed the submucosal, or Meissner’s plexus, and the myenteric, or Auerbach’s plexus (82, 185, 194). These two plexuses regulate major functions in the GI tract. Anatomically, the submucosal plexus is located between the muscularis mucosae and circular muscle layers in the GI tract (Fig.1.1). The submucosal plexus is the smaller of the two plexuses and consists of fewer neurons (82). It is primarily involved in controlling absorption, secretion, and local blow flood to the gut (174, 198, 3 206). In contrast, the myenteric plexus, the larger of the two plexuses, is located between the circular and longitudinal muscle layers of the GI tract (185)(Fig.1.1). The role of the myenteric plexus is primarily motility focused, as it is responsible for the coordinated contraction and relaxation within GI tract (89, 97, 182). This is an important process, as it allows for the proper transit of ingested food particles through the GI tract, a process known as peristalsis. The ENS develops from neural crest progenitors expressing gene markers Sox10, FoxD3, p75 neurotrophin receptor, Phox2b, RET, and endothelin B receptor (EDNRB) (32). As these progenitors differentiate into enteric neurons that populate the GI tract, they retain expression of Phox2b and RET, but lose expression of other progenitor cell markers. Enteric neurons also gain the expression of PGP9.5, β3-tubulin, and HuC-D (32). As they mature, enteric neurons further differentiate into different types of neurons and can be further classified by immunohistochemical labeling of neuronal markers and neurotransmitters they express. Classification of these enteric neurons falls into three major types: motor neurons (both excitatory and inhibitory), interneurons, and sensory neurons (49). Excitatory motor neurons elicit their excitatory effects primarily via release of acetylcholine, and to a lesser degree, takykinin proteins such as substance P (49, 61). These neurons can also be identified by their expression of calretinin and γ-aminobutyric acid. In contrast, inhibitory motor neurons primarily utilize nitric oxide as a neurotransmitter, along with vasoactive intestinal peptide (VIP), ATP, and carbon monoxide (48, 214). Interneurons (ascending and descending) present along the length of the GI tract responsible for reflex pathways utilize acetylcholine as a neurotransmitter, along with a combination of other markers such as serotonin, 4 somatostatin, nitric oxide, VIP, and calretinin (48). Finally, intrinsic sensory neurons utilize acetylcholine, calcitonin gene-related peptide, and tachykinins as neurotransmitters, and can be further identified via labeling for calbindin, calretinin, and isolectin B4 (48). 5 Figure 1.1: Arrangement of the ENS within the GI tract. The ENS is distributed within the walls of the GI tract in primarily two plexuses, the myenteric plexus and submucosal plexus. The submucosal plexus is located between the muscularis mucosae and circular muscle, while the myenteric plexus is located between the circular and longitudinal muscle layers. 6 Extrinsic inputs into the ENS Despite extensive intrinsic neuronal inputs into the ENS to control GI function, the ENS also receives extrinsic inputs from the CNS to modulate GI activity (128, 150). CNS inputs into the ENS consist of sympathetic, parasympathetic, and sensory innervation arranged in vagal, thoracolumbar, and craniosacral pathways (60). The sympathetic and parasympathetic inputs to the ENS can modulate GI function by stimulating GI motility and digestion with parasympathetic stimuli, and inhibiting GI motility and digestion with the sympathetic pathway (73, 128, 150). GI motility is modulated by parasympathetic efferent motoneurons that innervate the GI tract and are derived primarily from the dorsal motor nucleus of the vagal nerve for the upper regions of the GI tract, and preganglionic fibers from lumbosacral regions of the spinal cord in the distal regions of the colon (19). These preganglionic efferent motoneurons release acetylcholine to bind nicotinic receptors and activate interstitial cells of Cajal and neurons in the myenteric plexus to elicit muscle contraction in the GI tract (19, 168). However, vagal activation of non-adrenergic, non-cholinergic postganglionic pathways also allows for release of nitric oxide and vasoactive intestinal polypeptide in order for the relaxation of intestinal smooth muscle (19). In contrast, sympathetic modulation of GI activity originate from thoracolumbar regions of the spinal cord to several sympathetic ganglia in order to supply sympathetic innervation to the GI tract. Postganglionic fibers from the celiac ganglion supply sympathetic innervation to the proximal GI tract such as the stomach, while neurons from the superior mesenteric ganglion and inferior mesenteric ganglion provide sympathetic innervation to the small and large intestines, respectively (19). Unlike parasympathetic nerves, the preganglionic 7 sympathetic neurons primarily utilize norepinephrine as a neurotransmitter (19). Sympathetic modulation of intrinsic ENS neurons contributes to decreased motility in the GI tract, and increased tone in intestinal sphincters (106). In addition to modulating GI motility, sympathetic and parasympathetic nerves are distributed to lymphoid organs and immune tissues in the GI tract and have immunomodulatory effects on the GI system (24, 118, 186). Broadly speaking, parasympathetic activation is associated with anti-inflammatory effects in the gut, while sympathetic activation can lead to either pro- or anti- inflammatory effects, depending on levels of released neurotransmitters (16, 119, 186). Sympathetic signaling through α- adrenergic receptors can induce release of TNF-α from macrophages, promote Th2 T- lymphocyte activation, and increase Th17 T-lymphocyte activation through increased expression of IL-23 by antigen presenting cells (3, 91). In contrast, cholinergic signaling from parasympathetic pathways can act directly onto enteric neurons that interact with nearby macrophages. Release of acetylcholine by these enteric neurons can bind α-7- nicotinic acetylcholine receptors present on macrophages to inhibit the release of pro- inflammatory cytokine TNF-α (16). Interestingly, vagal efferent nerves can also interact with splenic sympathetic nerves within the celiac ganglion to elicit anti-inflammatory effects via the splenic nerve. Activation here leads to acetylcholine release in T- lymphocytes and thereby bind α-7-nicotinic acetylcholine receptors on splenic macrophages to inhibit release of TNF-α (16). In addition to parasympathetic and sympathetic nerves, even sensory neurons have immunomodulatory properties and can inhibit immune activation in neutrophils, monocytes, and macrophages (28). Therefore, the contributions from the CNS in 8 immune modulation play important roles in homeostasis and can contribute to intestinal inflammation, ENS damage, and the pathogenesis of IBD. ENS damage and disease Damage to the ENS can arise from a multitude of sources and can lead to neuronal death. Since the ENS regulates GI secretomotor function, injury to the ENS can manifest as symptoms such as constipation, diarrhea, nutrient malabsorption, or abdominal pain (60, 108). ENS dysfunction can be due to malformations congenitally, direct modifications to neuronal structure and function, inflammatory effects, exposure to chemicals, changes in microbiota, or as a part of normal aging (57, 71, 120, 166). In congenital ENS malformation, dysfunctions in the ENS arises due to inappropriate development of enteric neurons. For example, Hirschsprung’s disease is due to the improper migration of neural crest cells during gestation and leads to aganglionsis in the distal portions of the GI tract, primarily affecting the colon (191). Genetic abnormalities in multiple genes such as RET, endothelin receptor type B, and Sox10 contribute to the pathogenesis of Hirschsprung’s disease (17, 21, 148). Without the development of a proper ENS in sections of the gut, aganglionic regions lack enteric neurons and enteric glia. These regions do not have proper motility functions and typically cannot relax, leading to fecal stasis, bacterial overgrowth in the GI tract, and potentially lead to sepsis (120, 195). Diseases causing neuropathies, such as diabetes, can lead to ENS damage by modifying neurons and neuronal function. In diabetes, elevated glucose levels in the body leads to the covalent linkage of glucose molecules, termed glycation, of neurons. 9 This occurs through a process known as non-enzymatic glycation, which can hinder neuronal functions and trigger nerve degradation (93, 202). Furthermore, diabetes may also activate other pathways such as the polyol, protein kinase C, mitogen-activated protein kinase (MAPK), TNF-α, NF-κB, cyclooxygenase, interleukin (IL), Wnt, and autophagy pathways, among others, to elicit neuronal damage and pathogenesis (38). This can lead to a host of GI symptoms (42) and can contribute to motility disorders and gastroparesis seen in diabetes patients (38, 202). Enteric glial cells within the ENS can act to limit neuronal degradation (109), but diabetic changes in enteric glial cells, such as changes in glial morphology and number, can limit their ability to protect enteric neurons and further contribute to diabetes-associated ENS dysfunction (144, 153). Inflammation in the GI tract is a very common cause of enteric neuropathies, but the pathogenesis varies significantly between diseases. For example, inflammatory damage to the ENS can be caused by infection, such as in Chagas disease caused by Trypanasoma cruzi infection. The inflammatory response stimulated by this infection can lead to megacolon or megaesophagus due to destruction of ENS segments (81, 180). In IBD, pathogenesis is thought to be due to the inappropriate immune activation against the microbiota in genetically susceptible patients (29, 179, 210, 222). This can lead to inflammation in the GI tract, a hallmark of IBD. Inflammatory stress on the ENS is distributed differently in the two forms of IBD: Crohn’s disease and ulcerative colitis. While tissue inflammation is limited to the superficial layers of the colon in ulcerative colitis (225), Crohn’s disease can elicit inflammation anywhere along the length of the GI tract. In addition, inflammation in Crohn’s disease can affect the entire thickness of the GI tract and not only the superficial layers as in ulcerative colitis (225). Inflammatory 10 damage in the GI tract can lead to the reorganization, damage, and death of both enteric neurons and enteric glial cells, thereby contributing to a loss of GI secretomotor functions and symptoms seen in IBD patients. Inflammatory stress causes neuronal death through the activation of caspase- dependent apoptotic pathways in enteric neurons via activation of purinergic receptors and pannexin channels (63, 129). Neuronal death due to inflammation does not target specific subpopulations (129), but instead leads to a generalized decreased neuronal density in the ENS. However, inflammatory stress can also contribute to changes in neuronal behavior, such as the development of hyperactivity in intrinsic primary afferent neurons during, and after inflammation, thereby contributing to greater afterhyperpolarizing potential (104, 105, 129). The change in neuronal activity in these intrinsic primary afferent neurons suggest alterations in normal intestinal sensation and changes in motor reflexes that rely on these afferent nerves. In addition to alterations in neuron activity, IBD elicit changes in neurotransmitter expression in enteric neurons. For example, IBD patients have an increased proportion of neurons expressing substance P (134), neuropeptide Y (10), and changes in number of neurons expressing vasoactive intestinal polypeptide (10, 173). In addition to inflammatory stress, ENS exposure to certain chemicals in the body can lead to detrimental effects. For example, chemotherapy treatment has many effects on the GI tract, but chemotherapy-induced neuronal death in the ENS can lead to serious dysmotility disorders (2, 43, 121, 208). Furthermore, chemical-induced ENS damage and alterations in neuronal and glial function can persist for extended periods of time, such as in Gulf War veterans exposed to pyridostigmine bromide decades ago 11 (70, 71). In this way, chemical damage to the ENS can have lasting effects even after offending chemicals are removed. The interplay between the ENS and the microbiota in the GI tract also has significant implications on GI disease and secretomotor function. Many groups have shown interactions between the microbiota, the ENS, and the CNS (35, 171, 204). Changes in the microbiota composition has profound effects on GI function, and GI diseases such as IBD have been linked with intestinal dysbiosis and alterations in microbiota (1, 139). Due to the complex nature of microbiota interactions, the specific effects due to changes in microbe distribution are not completely known and is a topic of study. Finally, the process of normal aging causes increased neurodegeneration overall, but neurons in the ENS are especially susceptible (166). Despite the plasticity of the ENS and its capability of neuronal outgrowth, aging leads to the reduction in the number of neurons and glia in the ENS (166). Furthermore, aging causes alterations in neuronal morphology and arrangement, suggesting alterations in neuronal behavior and function, and may contribute to GI motility disorders seen in old age, such as fecal incontinence and constipation (26, 52). Taken together, ENS neuropathy can be due to numerous reasons ranging from congenital malformations, direct neuronal modifications, inflammation, the exposure to chemicals, changes in microbiota, just from normal aging, or a combination of any of these factors. ENS neuropathy, irrelevant of cause, leads to GI secretomotor dysfunction by causing damage to both enteric neurons and enteric glia, thereby disrupting the ENS network. 12 Enteric Glia Enteric glia, together with enteric neurons, form the ENS. They are a type of non- myelinating peripheral glia that share similar characteristics as astrocytes of the CNS (50, 55, 84). They are in extremely close contact with each other (15) (Fig.1.2), and were historically thought of being merely a ‘support’ cell to enteric neurons by providing nutritional and structural support to the ENS. However, enteric glia have since been shown to have an important impact on enteric neurogenesis, activity, and survival (32, 65, 135). Enteric glia are derived from neural crest cells and begin development and colonization of the intestinal tract within the first two weeks in utero (55, 88, 163). During development, enteric glia express markers such as Sox10, FoxD3, B-FABP, glial fibrillary acidic protein (GFAP), and S100β, making them a distinct population compared to developing enteric neurons or neural crest progenitor cells (32). Therefore, current identification of enteric glia within the ENS is achieved by the labeling of glial-specific markers S100β and GFAP (32, 135, 164), and by morphology, as they have distinct astrocyte-like star shaped morphology with long and highly branched processes (50, 164, 165) (Fig.1.2). However, despite their similarities with glial populations in the CNS, enteric glia are a distinct population. This is evidenced by their embryonic origins from the neural crest, in opposed to astrocytes being derived from the neural tube (8, 55). Due to the differences in embryologic origin, enteric glia also express the Sox10 transcription factor, unlike astrocytes (127). 13 Figure 1.2: Enteric glial proximity to enteric neurons. Immunofluorescent labeling of enteric neurons with HuC/D (red) and enteric glia with glial-specific marker GFAP (grey). Enlarged area further demonstrates the proximity of glial processes to enteric neurons. Scale bar represents 50µm. 14 In the established ENS, enteric glia communicate with enteric neurons and other cell types such as intestinal epithelium to play an active role in GI secretomotor function. To interact with neuronal signaling, enteric glia express receptors for numerous neurotransmitters such as cholinergic receptors (27, 36), adrenergic receptors (62, 132), purinergic receptors (58, 64, 92), serotonin receptors (14), among others (130, 175, 224). This allows for enteric neurons to respond to nearby neuronal activity and be targets of innervation. In addition, enteric glia can also modulate neuronal signals in the ENS, thereby contributing to the control of GI motility (32, 65, 135) (Fig.1.3). In addition to interactions with enteric neurons, enteric glia can interact with non-neuronal cell types as well. Their interactions with intestinal epithelial cells play a role in the control of intestinal secretion and barrier function (110, 111, 170), and their ability to secrete and respond to cytokines can affect immune activity (90, 162) (Fig.1.3). A more in-depth examination of enteric glial cell characteristics that support their role in immune modulation is presented in chapter 2 of this dissertation. Taken together, enteric glia play an active role in normal ENS signaling as well as secretomotor and immune modulation activities in the GI tract. 15 Figure 1.3: Overview of enteric glial functions. Enteric glia play an active role in the communication and interaction between enteric neurons, enterocytes, and immune cells in the intestinal tract. These interactions are through neurotransmitters and soluble molecules such as nitric oxide or secreted cytokines in order to affect GI secretomotor function, barrier function, and immune activity. 16 In addition to their role in ENS signaling, enteric glia also play a role in neurogenesis and neuronal maintenance in the ENS. Studies demonstrate that enteric glia help maintain the health of the ENS and recover from injury via the expression of neurotrophic factors that can stimulate neuron outgrowth and regeneration (11, 66, 181). However, during oxidative stress and neuronal damage, enteric glia can also play the role of executioner and contribute to neuronal death (18). In mice, there are roughly equal numbers of enteric glia and enteric neurons in the GI tract, but the ratio between glia and neurons increases in higher order animals, reaching roughly seven-to-one enteric glia-to-neuron ratios in humans (51, 74). This suggests that enteric glia can effectively monitor neuronal health and function, and that increasing numbers of enteric glia are necessary for the increasing complexity of GI functions in higher organisms. The Immune System The ENS in the walls of the GI tract does not exist in isolation. In fact, many immune cells are in the surrounding microenvironment of both the submucosal and myenteric plexus. Dendritic cells, muscularis macrophages and memory T-lymphocytes are all normally distributed within the mucosa, submucosa, and muscular layers of the GI tract (5, 33) to monitor the local microenvironment for any potential pathogens (Fig.1.4). The proximity of immune cells to the ENS lends to interactions between cells of the ENS and the immune system (138, 209). While having immune cells in close proximity to the ENS is beneficial in eliminating nearby pathogens, immune activation or dysregulation can theoretically have immediate effects on the ENS. 17 Figure 1.4: Overview of typical GI immunity. Spread within the mucosa, submucosa, and muscular layers of the GI tract are dendritic cells, lymphocytes, and macrophages. Nearby GI-associated lymphoid tissues (GALT) and regional mesenteric lymph nodes contain additional immune cells to respond to pathogens. In the submucosal and myenteric plexuses, muscularis macrophages are present in close proximity to the ganglia. 18 Broadly, the immune system is responsible for host defense and is involved in the recognition and removal of pathogens, foreign molecules, and damaged cells and tissues. It is divided into two main components, the innate and adaptive immune systems. The innate immune system is responsible for a rapid response to an invading pathogen by limiting its spread, phagocytosing and destroying pathogens, and communicate with the adaptive immune system to mount an additional response to a given pathogen (169). Innate immunity relies on molecular patterns common on many pathogens to generate a generic immune response (83, 145). In contrast, the adaptive immune system does not respond with the same speed as the innate immune system and relies on activity in the innate immune system to convey antigens to direct it to the proper target. In addition, the adaptive immune system has the ability to generate a ‘memory’ of a previous immunologic insult such that subsequent exposure to the same insult elicits a stronger and more rapid adaptive immune response (145, 169). Activation in either the innate and adaptive immune systems can lead to inflammation within the ENS, contributing to the pathogenesis of ENS disorders and IBD. Currently, numerous therapeutics have been developed to target specific steps in each arms of the immune system (75), and there continues to be rigorous study into immune regulation as a means to limit inflammatory damage to the ENS. Inflammation Inflammation has been recognized since ancient Roman times. The cardinal signs of inflammation were described by Aulus Cornelius Celsus as being rubor (redness), tumor (swelling), calor (heat), and dolor (pain) (193). The process of 19 inflammation represents a broad response to limit the spread of pathogens and deliver immune cells, among other products, to help eliminate threats. However, inflammation is also a major driver for neuroplastic changes in the ENS (207). Changes to neuron survival and the overall ENS network leads to functional alterations in the ENS and contributes to secretomotor symptoms in diseases such as IBD. The overall inflammatory process is accomplished by first increasing blood flow and vascular permeability to the site of infection or injury. During infection, the increase in fluid to the area helps to isolate a pathogen to prevent spread (122). Furthermore, the increase in blood and lymph flow to the area helps bring leukocytes to the site of injury or infection to help progress the immune reaction (122). Initial immune cells recruited to the inflamed area primarily consists of neutrophils and monocytes, but progressively expands to include macrophages and T-lymphocytes over time (122, 152). If the recruited immune cells are incapable of clearing the pathogen, granulomas may form to sequester and isolate the offending agent from spreading to other regions of the tissue or body (122). The inflammatory pathway resolves once the pathogen or inflammatory agent is cleared from the body, and tissue repair and regeneration is activated to reverse damage incurred by the inflammatory process. One of the ways immune cells carry about effector functions is through the expression of cytokine molecules. Cytokines, which are soluble chemicals in the bloodstream used for cell signaling, and chemokines, which are chemotactic cytokines, contribute either to the development or resolution of inflammation, depending on the molecule (41, 78). They are expressed by both the innate and adaptive arms of the immune system, and the expression and levels of different cytokines are relevant in the 20 pathogenesis of inflammatory diseases such as IBD. They cytokine landscape in IBD is very diverse and includes a multitude of pro-inflammatory and anti-inflammatory cytokines and chemokines in its pathogenesis (47, 136, 199). Among them, therapeutics have been developed and are being tested to neutralize a portion of those cytokines and chemokines, such as Fontolizumab targeting IFNγ, Infliximab (tumor necrosis factor), Tocilizumab (IL-6), Ustekinumab and briakinumab (IL-12 and IL-23), Anrukinzumab (IL-13), Secukinumab (IL-17), Vercirnon (chemokine receptor CCR9), Eldelumab (CXCL10), and Smad7 anti-sense oligonucleotides (CCL20) (47, 136, 199). Furthermore, the therapeutic administration of recombinant IFNβ, recombinant IL-10, or recombinant IL-11 is also being studied as a means to limit inflammatory burden in IBD (136). Therefore, the regulation of cytokine and chemokine levels is an important area to regulate inflammation and is the current strategy to limit inflammatory damage to the ENS and surrounding tissue in IBD. The inflammatory pathway represents a destructive process as recruited neutrophils and monocytes phagocytose and destroy and clear foreign pathogens and cellular debris. However, their destructive nature and release of reactive oxygen species can be non-discriminant and cause collateral damage to the surrounding tissue (124, 142, 152). The prolonged activation of inflammation in a tissue can lead to fibrosis and scar formation, leading to the replacement of damaged tissue with non-functional scar tissue. In the context of GI inflammation, intestinal fibrosis presents a very serious complication in IBD patients. The combination of inflammation-induced neuron loss in the ENS and increased intestinal fibrosis leads to GI secretomotor dysfunction in IBD patients and leads to an increased morbidity and mortality (76, 99). 21 Despite its destructive properties, the process of inflammation can be beneficial to tissue repair and regeneration. Inflammatory recruitment of immune cells, especially macrophages, play a major role in wound healing and tissue regeneration. Specifically, M2-subtype macrophages function to remove neutrophils and apoptotic cellular debris from inflammatory regions, to promote angiogenesis, extracellular matrix synthesis, stimulate progenitor cell activation, and fibroblast proliferation (80, 94, 142). Furthermore, at the resolution of inflammatory processes, the release of mediators such as resolvins by endothelial cells inhibits neutrophil recruitment and extravasation to damaged tissue, and stimulates macrophages to clear apoptotic neutrophils and other cell debris from the inflammatory site (46, 176). Many of these inflammatory functions, especially during the early stages of inflammation, are carried out by the innate immune system, and this arm of the immune system represents a major player in the GI inflammation. The Innate Immune System The innate immune system represents one of the first lines of defense against infectious agents and pathogens, the first line of defense being epithelial barriers that separate our bodies from the external environment and potential pathogens. The innate immune system consists of fast responding immune cells such as dendritic cells, macrophages, polymorphonuclear cells (including neutrophils, basophils, and eosinophils), and mast cells (83). These cells are positioned in areas of close proximity with potential pathogens, such as the mucosal layer of the intestinal tract. Innate immune cells recognize pathogens by common molecular patterns expressed on their cell surface, termed pathogen-associated molecular patterns (67, 83). Binding of these 22 pathogen-associated molecular patterns onto ‘pattern recognition receptors’ expressed on immune cells activates the immune cell, leading to phagocytosis and clearing of the pathogen (67, 83). Activation of innate immune cells such as dendritic cells can also lead to the migration of the dendritic cell to nearby lymph nodes to stimulate and activate the adaptive immune system. Depending on the severity of pathogen invasion, tissue inflammation may also ensue. Pattern recognition receptors are divided into 3 major types: Toll-like receptors (TLR), NOD-like receptors, and RIG-I-like receptors (96). These pattern recognition receptors bind pathogen-associated molecular patterns typically consisting of bacterial, viral, or fungal components. However, pattern recognition receptors can also bind molecules that represent cellular damage, termed damage-associated molecular patterns (59). For example, TLR1&2 recognize bacterial cell wall components like peptidoglycan, and triacyl lipopeptides, TLR3 recognize dsRNA molecules, TLR4 recognize lipopolysaccharide (LPS), TLR5 recognize flagellin molecules, TLR7&8 recognize ssRNA molecules, and TLR9 recognize dsDNA (83, 96, 226). RIG-I-like receptors recognize different viral pathogens, while NOD-like receptors a variety of bacterial, viral, and fungal pathogen-associated molecular patterns, along with non- microbial damage-associated molecular patterns (96, 226). However, binding onto pattern recognition receptors does not always elicit the destruction of the microorganism. Pattern recognition receptors have also been shown as a means to monitor and recognize commensal microorganisms in the microbiota (23, 95). Binding of TLR molecules by commensal organisms is necessary for intestinal homeostasis, is a way to modulate microbiota populations, can induce anti-inflammatory responses in the 23 intestines, and modulates intestinal epithelial metabolism (23, 30, 95, 154, 218). In the context of the ENS, enteric glial cells can express pattern recognition receptors such as TLR4 to detect LPS molecules, and can release nitric oxide via TLR4 binding (107, 201). As described above, the activation of innate immune cells can trigger phagocytosis of potential pathogens. The process of phagocytosis represents an important pathway in innate immune cells to both clear pathogens and to prepare antigens for presentation to the adaptive immune system (67). Once recognized by innate immune cells, restructuring of the cellular cytoskeleton allows for the engulfment and internalization of foreign molecules into phagosomes (44). The maturation of the phagosome into a phagolysosome involves the fusion with lysosomes, the acidification of vesicle contents, and the accumulation of reactive oxygen species and hydrolases including various proteases, lipases, nucleases, glycosidases, and phosphatases to degrade internalized particles (44, 203). In the GI tract, dendritic cells resident macrophages, and recruited neutrophils and monocytes are the primary cell populations carrying out phagocytosis activities. Antigen Presenting Cells While phagocytosis in neutrophils is primarily to clear pathogens and not to present antigens to the adaptive immune system, dendritic cells and macrophages play an additional role as professional antigen presenting cells to load phagocytosed particles onto major histocompatibility complex (MHC) II molecules for presentation to the adaptive immune system (145, 183). Antigen presenting cells form an important transition between the innate and adaptive immune system and can direct an immune 24 response towards specific targets. In addition to their phagocytic ability and constitutive expression of MHC II molecules, professional antigen presenting cells are also capable binding and activating T-lymphocytes (145, 169). This is accomplished by the expression of T-lymphocyte co-stimulatory molecules such as CD40, CD80, and CD86, along with activating cytokines to stimulate T-lymphocyte proliferation and differentiation (169). Although antigen presentation to T-lymphocytes is primarily carried out by dendritic cells and macrophages as professional antigen presenting cells, growing evidence supports the presence of ‘atypical’ antigen presenting cells as well (87, 137, 183). These atypical antigen presenting cells are characterized by the ability to express MHC II molecules in an inducible manner, as opposed to constitutive expression of MHC II in professional antigen presenting cells (87, 183). Furthermore, although atypical antigen presenting cells can express co-stimulatory molecules, they are generally not as effective as professional antigen presenting cells at triggering T- lymphocyte activation, differentiation, and proliferation (87, 177). In fact, there are conflicting studies debating whether atypical antigen presenting cells are functional at inducing T-lymphocyte activation (87, 157, 177). Currently, cells such as mast cells, basophils, eosinophils, and type 3 innate lymphoid cells have been classified as atypical antigen presenting cells (20, 40, 45, 69, 87, 112, 141, 151, 220). However, due to conflicting findings on their capabilities to effectively function as an antigen presenting cell, the true role of atypical antigen presenting cells remains to be elucidated. Within the ENS, enteric glial cells show promise to also act as an atypical antigen presenting cell. Enteric glia have been shown to express MHC II molecules, T-cell co- 25 stimulatory molecules, and can interact activate T-lymphocytes in vitro (4, 53, 201). Moreover, astrocyte cell populations in the CNS that are related to enteric glia show phagocytic capabilities and can uptake (85). A more detailed examination into the characteristics of enteric glial cells that supports their potential role as an immune modulator is presented in chapter 2 of this dissertation. Type II Major Histocompatibility Complex As described above, the MHC II molecule is constitutively expressed on professional antigen and functions as a crucial component by which self-derived or phagocytosed antigens are presented on the cell surface to T-lymphocytes (145, 183). The identity of presented peptides activates specific T-lymphocytes, and therefore modulates immune activation. Slight changes in the peptides presented can drastically change the resultant immune reaction, as simple post-translational modification in presented peptides can change a non-existent immune response into a complete autoimmune cascade (13, 72). Inappropriate peptide loading onto MHC II molecules and dysfunctional antigen presentation are contributing factors in the development of multiple diseases, such as IBD, systemic lupus erythematosus, and multiple sclerosis (25, 113, 123, 211, 216). Therefore, the proper loading of peptides onto MHC II molecules is crucial in directing appropriate immune responses. In the mouse (the animal model used in this dissertation), the MHC II molecule is a heterodimer composed of α1, α2, β1 and β2 chains (77, 184, 189) (Fig.1.5a). These are expressed on two functional α loci (Aa and Ea), and two functional β loci (Ab and Eb) on chromosome 17 (77, 189) (Fig.1.5b). Gene products from either the Aa and Ab pair or Ea and Eb pair combine to form functional MHC II molecules. 26 Figure 1.5: Murine MHC II. (A) The MHC II molecule is a heterodimer composed of the α1-α2 peptide dimerized with the β1-β2 peptide, forming a peptide binding groove to present peptide fragments to the adaptive immune system. (B) The MHC II molecule is expressed from two functional α loci (Aa and Ea) and two functional β loci (Ab and Eb). Gene products from either Aa and Ab or Ea and Eb combine to form the MHC II molecule. 27 Newly synthesized MHC II molecules from the endoplasmic reticulum are attached with CD74, also known as the invariant chain (158, 190). The attachment of the invariant chain serves two major purposes: 1) to occupy the peptide binding groove on MHC II molecules and prevent inappropriate peptide binding, and 2) to guide newly synthesized MHC II to low-pH endosome compartments (158, 190). As MHC II molecules mature, the invariant chain is degraded until only a small peptide, the class II- associated invariant-chain peptide (CLIP), remains bound to the MHC II peptide binding groove. It is not until the fusion with phagolysosomes that CLIP molecules are displaced from the peptide binding groove via the activity of the H2-M enzyme (158), and MHC II molecules are free to load antigens (Fig.1.6). Loaded MHC II molecules are transported to the cell surface where it can then, in conjunction with other co-stimulatory molecules, bind and activate T-lymphocytes. Although peptide loading onto MHC II molecules are generally derived from extracellular antigens that have been phagocytosed and processed, atypical loading of MHC II molecules can also occur through the loading of intracellular peptides via the autophagy pathway (37, 187). Despite loading intracellular antigens onto MHC II molecules, autophagy-induced peptide loading onto MHC II can not only avoid the development of auto-immunity but can also elicit an anti-inflammatory effect by limiting T-lymphocyte activation (212). 28 Figure 1.6: Maturation of MHC II molecules. Newly synthesized MHC II molecules are bound with the invariant chain peptide and transported into phagolysosomes, where proteolytic cleavage processes the bound invariant chain into the CLIP peptide while maintaining MHC II binding. The association of MHC II with the H2-M molecule (the murine equivalent to the human DM molecule) allows for the release of the CLIP 29 Figure 1.6 (cont’d): peptide from the MHC II peptide binding groove in order to load endocytosed peptides for presentation on the cell surface. Processing of cytoplasmic proteins in autophagolysosomes can also merge into this pathway and load cytoplasmic peptides onto MHC II molecules for presentation. 30 The autophagy pathway is important during development, but it also represents a method for cells to degrade and recycle cellular components during times of stress or starvation (102, 146). The autophagy pathway also plays a role in pathogen defense by playing a role in the degradation and presentation of intracellular bacterial and viral pathogens (103, 131). Furthermore, in a normal resting state, basal activation of autophagy and loading of autophagy-derived intracellular peptides contributes to immune tolerance (156). In the context of GI homeostasis, autophagic pathways are important in the maintenance of proper microbiota composition (200, 217), as autophagy dysfunction is associated with intestinal dysbiosis (98). The regulation of the autophagy pathways is an important method of maintaining homeostasis, and defective autophagy pathways have far reaching effects in the body. For example, dysfunction in the autophagy pathway is associated with genetic instability and tumorigenesis (101, 117), neurodegenerative diseases such as Huntington’s disease and Parkinson’s disease (115, 156, 213), GI diseases such as IBD and liver dysfunction (79, 147), and myopathies in both cardiac and skeletal muscle (140, 196). In the context of antigen presentation, while basal activation of autophagy-mediated MHC II loading is beneficial, a dysregulated autophagy pathway could theoretically lead to the presentation of inappropriate antigens to the adaptive immune system, but this has yet to be established (34, 172, 187, 205). The Adaptive Immune System The adaptive immune system demonstrates fairly different characteristics compared to the innate immune system. While innate immunity occurs quickly after binding pattern recognition receptors, the activation of the adaptive immune system is 31 much slower and generally involves cellular proliferation of specific adaptive immune cells after activation (215). This represents a second ‘phase’ of immunity after the innate immune system. A factor contributing to response time is the fact that innate immune cells are positioned in areas near potential infection sites. However, the adaptive immune system is generally positioned in different lymphoid organs and require recruitment (145). As described above, the adaptive immune system also requires contributions from the adaptive immune system in the form of antigen presentation cells. The presentation of antigens on MHC II molecules is crucial in order to direct the adaptive immune response (145, 183). In contrast, the activation of the innate immune system by binding of pattern recognition receptors elicits a consistent innate response, and not a ‘tailored’ adaptive response to specific antigens. Finally, unlike the innate immune system which provides the same response at each exposure to a given antigen, the adaptive immune system provides a lasting ‘memory’ for a quicker and more robust reaction at subsequent exposures to the same antigen. The adaptive immune system is comprised of two major cell types: T- lymphocytes and B-lymphocytes. While B-lymphocytes are associated with humoral immunity from antibodies secreted by activated B-lymphocytes, T-lymphocytes are associated with cell-mediated immunity via direct cell-to-cell interactions. These two cell types do not act independently from each other, as specific T-lymphocyte subsets are involved in the activation of B-lymphocytes as well (12, 145). Contributions from both B and T-lymphocytes leads to robust adaptive immune responses. 32 T-lymphocytes T-lymphocytes form in the thymus, and through rigorous selection processes, they develop into either CD4-expressing or CD8-expressing T-lymphocytes (54). Once developed, but prior to encountering an antigen or stimulation, they are in a naïve and inactive state stored in lymphoid organs (145). However, antigen presentation and stimulation from antigen presenting cells causes the activation and proliferation of T- lymphocytes. The activation state of a T-lymphocyte can be determined not only by the cytokines it expresses and proliferation rate, but also by the cell surface markers that are present. For example, acute markers for T-lymphocyte activation include CD69, which becomes detectable on the cell surface within several hours, and CD25, which becomes detectable within several days (31, 178). Once activated, T-lymphocytes take on effector roles to bring about immune responses (145). CD8-expressing T-lymphocytes are also known as cytotoxic T-lymphocytes, and they carry out their cytotoxic effector functions via cell-to-cell contact and binding of ‘non-self’ MHC I molecules (145). These T-lymphocytes are especially important in the detection of intracellular pathogens such as viruses and modified ‘self’, such as in cancer (159). CD8+ T-lymphocytes rely on the recognition and binding of cell surface receptors and perform cytotoxic effects by perforating the target cell wall with perforin. The subsequent introduction of granzyme molecules into the target cell cytoplasm activates the caspase cascade and triggers apoptosis in the target cell (145). Unlike their CD8-expressing counterparts, CD4+ T-lymphocytes do not generally carry out their effector functions via cell receptor binding. Instead, their effector functions are carried out by releasing soluble molecules and cytokines (145). CD4+ T- 33 lymphocytes are also referred to as helper T-lymphocytes due to their function to aid in the activation of other T-lymphocyte subtypes or other immune cells. CD4+ T- lymphocytes can be further subdivided into different cell types based on the primary cytokine or transcription factor expressed by each CD4+ T-lymphocyte subtype. T- lymphocyte subtypes that are of interest in this dissertation include Th1, Th2, Th17, and Treg T-lymphocytes. Each T-lymphocyte subtype expresses a unique combination of cytokines and can drive different forms of immune reactions. For example, Th1 T-lymphocytes are identified by their expression of IFNγ, IL-2, and TNF-β to activate macrophages, B cells, and cell mediated immunity with a predominantly phagocytic-dependent inflammation (161). Crohn’s disease has traditionally been thought of as a Th1-mediated disease (161), but recent studies demonstrate a joint role for Th1 and Th17 in its pathogenesis (133). In contrast, ulcerative colitis is regarded as a Th2-mediated disease with distinct cytokine profiles, expressing IL-4, IL-5, IL-6, IL-9, IL-10, and IL-13 (133, 160, 161, 188). Th2 T-lymphocytes can also inhibit Th1 effects such as phagocyte-dependent inflammation, while inducing an antibody-mediated response (161). Depending on the surrounding environment, activated Th17 T-lymphocytes express multiple cytokines including IL-9, IL-10, IL-17A, IL-17F, IL-21, IL-22, granulocyte-macrophage colony stimulating factor, and IFNγ (9, 116). The physiologic role for Th17 T-lymphocytes is to regulate neutrophil recruitment and provide immunity against bacterial and fungal pathogens via cytokine and chemokine release (9). However, improper Th17 regulation contributes to IBD pathogenesis and is associated with auto-immune diseases such as type 1 diabetes, rheumatoid arthritis, systemic lupus erythematosus and multiple 34 sclerosis (9, 100, 197). Finally, the Treg subtype of T-lymphocytes act in an anti- inflammatory manner and limits proliferation in the other T-lymphocyte subtypes to promote immune tolerance, limit auto-immunity, and maintain tissue homeostasis and wound repair (86, 167, 221). They can be identified through their expression of the FoxP3 transcription factor, and they carry out immunosuppressive activities through the release of anti-inflammatory cytokines IL-10 and transforming growth factor beta (TGF- β), sequestering pro-inflammatory cytokines, and inducing the anergy and apoptosis of other pro-inflammatory T-lymphocyte subtypes (86, 100, 167). Current models of intestinal inflammation A variety of animal models have been developed in order to model intestinal inflammation and inflammatory bowel diseases. These models are primarily chemically induced, genetically induced, or cell-transfer induced colitis models. While many different genetic models have been developed that develop intestinal inflammation, these models generally also trigger inflammatory responses or pathology in other organ systems as well. One of the more widely used animal knockouts are IL-10 knockout animals that demonstrate a spontaneous development of colitis after roughly 3 months of age (125, 126). Other knockout models include IL-2 knockout, TGFβ knockout, Toll- Like Receptor 5 knockout, signal transducer and activator of transcription 3 knockout, T- lymphocyte receptor α knockout, among others (125, 126). Adoptive transfer of CD4+ or CD8+ T-lymphocytes is another technique to model intestinal inflammation. Naïve CD4+ T-lymphocytes transferred from the spleen of wildtype animals to immune-deficient mice cause a Th1-mediated response to elicit transmural colitis when in the absence of regulatory T-lymphocytes (125). Similarly, transfer of naïve CD8+ T-lymphocytes to 35 recombination activating gene-2 knockout animals elicits colitis via activated CD8+ T- lymphocytes expressing IL-17. In addition to the transfer of naïve T-lymphocytes, the transfer of CD8+ T-lymphocytes that are specific for heat shock protein 60 or ovalbumin (in the presence of administered ovalbumin) also elicits intestinal inflammation (125). Despite the effectiveness of genetic models or adoptive transfer of T-lymphocytes in generating intestinal inflammation, the most common type of model utilized for IBD comes in the form of chemically induced intestinal inflammation and IBD, due to the low cost and the ease of these models (155). These chemicals include dextran sulfate sodium (DSS), oxazolone, dinitrobenzene sulfonic acid, trinitrobenzene sulfonic acid, and acetic acid. These models are beneficial in that it does not require specific genetic strains of animals and can be administered to form discrete regions of intestinal inflammation. For example, DSS-induced colitis utilizes 1-5% of the polymer DSS in drinking water to induce acute phase colitis, characterized by increased levels of TNF-α (125, 155). Since colitis is triggered by intestinal epithelial damage due to osmotic stress from DSS and not due to the activation of adaptive immune cells, colitis can be elicited in animals lacking T- and B- lymphocytes (39). In contrast, oxazolone-induced colitis utilizes an intrarectal administration of haptenizing agent oxazolone with ethanol to elicit an acute development of colonic inflammation. As oxazolone is a haptenizing agent, it requires an intact adaptive immune system, specifically working via natural killer T cells (68). Similarly, the administration of haptenizing agents dinitrobenzene sulfonic acid and trinitrobenzene sulfonic acid by enema with ethanol allows for the development of regional intestinal inflammation and favors a Th1-mediated inflammatory response (125, 155). Finally, acetic acid enema causes intestinal epithelial damage and 36 is characterized by neutrophil recruitment and ulceration and necrosis in the mucosal and submucosal layers (155). This dissertation aims to examine the contributions of enteric glia on the development of intestinal inflammation. Given the severity of inflammation in the developed IBD models described above, experiments in this dissertation utilize a different form of inflammatory stimuli. Based on findings from Zhang et al. (223), we utilize intraperitoneal (IP) injections of IFNγ and LPS as inflammatory stimuli. This is to ensure that inflammatory stress does not overwhelm our experimental model and to prevent inflammatory responses being dominated by other immune cell types, thereby masking contributions from enteric glia. Details of our model and injection of inflammatory stimuli are described in Chapter 3. Summary and Aims of Dissertation The ENS is composed of enteric neurons and enteric glia, and it is the primary regulator of GI function (48, 194). Inflammatory damage to the ENS can lead to the loss of enteric neurons and glia, contributing to secretomotor dysfunctions seen in the GI diseases (56). Therefore, immune modulation in the GI tract is extremely important in maintaining ENS health and function. Neuroimmune interactions are becoming increasingly recognized as important aspects to immune modulation. As a part of the ENS, enteric glia demonstrate similar characteristics to antigen presenting cells. This suggests that enteric glia may play a similar role as antigen presenting cells and interact with the adaptive immune system, but their exact role in immune modulation has yet to be studied. 37 During homeostasis, enteric glia play important roles in the maintenance of enteric neuronal health and can modulate neuronal activity to maintain normal GI secretomotor functioning (32, 65, 135). However, during inflammation, enteric glia can contribute to neuronal death and express MHC II and T-lymphocyte co-stimulatory molecules (4, 18, 53, 201). Co-culture experiments demonstrate the ability for enteric glia to also modulate T-lymphocyte activity (90). The objective of this dissertation is to understand the role enteric glia play in vivo in modulating immune activity during homeostasis and in pro-inflammatory environments. Chapter 2 takes a further look into recent studies that demonstrate the potential for enteric glia to function as an immunomodulator. Chapter 3 investigates the capabilities of enteric glia to act as an antigen presenting cell, their effects on T- lymphocyte activation, and the contributions autophagy has on glial MHC II expression. Together, these chapters aim to improve the understanding of how enteric glia interact with T-lymphocytes, and their role in immunomodulation. 38 REFERENCES 39 REFERENCES 1. Ananthakrishnan AN, Bernstein CN, Iliopoulos D, Macpherson A, Neurath MF, Ali RAR, Vavricka SR, Fiocchi C. Environmental triggers in IBD: A review of progress and evidence. Nat. Rev. Gastroenterol. Hepatol. 15 Nature Publishing Group: 39–49, 2018. 2. Appelbaum J, Wells D, Hiatt JB, Steinbach G, Stewart FM, Thomas H, Nghiem P, Kapur RP, Thompson JA, Bhatia S. Fatal enteric plexus neuropathy after one dose of ipilimumab plus nivolumab: A case report. J Immunother Cancer 6, 2018. 3. Bai A, Lu N, Guo Y, Chen J, Liu Z. Modulation of inflammatory response via α2- adrenoceptor blockade in acute murine colitis. Clin Exp Immunol 156: 353–362, 2009. 4. Barcelos Morais Da Silveira A, De Oliveira EC, Neto SG, Luquetti AO, Toshio Fujiwara R, Correa Oliveira R, Brehmer A, Barcelos A, Oliveira EC De, Neto SG, Luquetti AO, Toshio R, Correa R, Brehmer A. Enteroglial cells act as antigen-presenting cells in chagasic megacolon. Hum Pathol 42: 522–532, 2011. 5. Bartolomé-Casado R, Landsverk OJB, Chauhan SK, Richter L, Phung D, Greiff V, Risnes LF, Yao Y, Neumann RS, Yaqub S, Øyen O, Horneland R, Aandahl EM, Paulsen V, Sollid LM, Qiao SW, Baekkevold ES, Jahnsen FL. Resident memory CD8 T cells persist for years in human small intestine. J Exp Med 216: 2412–2426, 2019. 6. Bayliss WM, Starling EH. The movements and innervation of the small intestine. J Physiol 24: 99–143, 1899. 7. Bayliss WM, Starling EH. The movements and the innervation of the large intestine. J Physiol 26: 107–18, 1900. 8. Bayraktar OA, Fuentealba LC, Alvarez-Buylla A, Rowitch DH. Astrocyte development and heterogeneity. Cold Spring Harb Perspect Biol 7, 2015. 9. Bedoya SK, Lam B, Lau K, Larkin J. Th17 cells in immunity and autoimmunity. Clin. Dev. Immunol. 2013 Hindawi Limited: 986789, 2013. 10. Belai A, Boulos PB, Robson T, Burnstock G. Neurochemical coding in the small intestine of patients with Crohn’s disease. Gut 40: 767–774, 1997. 11. Le Berre-Scoul C, Chevalier J, Oleynikova E, Cossais F, Talon S, Neunlist M, Boudin H. A novel enteric neuron-glia coculture system reveals the role of glia in neuronal development. J Physiol 1542: 33–36, 2016. 12. Biram A, Davidzohn N, Shulman Z. T cell interactions with B cells during 40 germinal center formation, a three‐step model. Immunol Rev 288: 37–48, 2019. 13. Birnboim HC, Lemay A-M, Lam DKY, Goldstein R, Webb JR. Cutting edge: MHC class II-restricted peptides containing the inflammation-associated marker 3- nitrotyrosine evade central tolerance and elicit a robust cell-mediated immune response. J Immunol 171: 528–32, 2003. 14. Boesmans W, Cirillo C, Van den Abbeel V, Van den Haute C, Depoortere I, Tack J, Vanden Berghe P. Neurotransmitters involved in fast excitatory neurotransmission directly activate enteric glial cells. Neurogastroenterol Motil 25, 2013. 15. Boesmans W, Martens MA, Weltens N, Hao MM, Tack J, Cirillo C, Vanden Berghe P. Imaging neuron-glia interactions in the enteric nervous system. Front Cell Neurosci 7: 183, 2013. 16. Bonaz, Bruno; Sinniger, Valerie; Pellissier S. Anti-inflammatory properties of the vagus nerve: potential therapeuitc implications of vagus nerve stimulation. J Physiol 82: 1496–1514, 2011. 17. Brosens E, Burns AJ, Brooks AS, Matera I, Borrego S, Ceccherini I, Tam PK, García-Barceló M-M, Thapar N, Benninga MA, Hofstra RMW, Alves MM. Genetics of enteric neuropathies. Dev Biol 417: 198–208, 2016. 18. Brown IAM, McClain JL, Watson RE, Patel BA, Gulbransen BD. Enteric Glia Mediate Neuron Death in Colitis Through Purinergic Pathways That Require Connexin-43 and Nitric Oxide. C Cell Mol Gastroenterol Hepatol 2: 77–91, 2016. 19. Browning KN, Travagli RA. Central nervous system control of gastrointestinal motility and secretion and modulation of gastrointestinal functions. Compr Physiol 4: 1339–1368, 2014. 20. Von Burg N, Chappaz S, Baerenwaldt A, Horvath E, Bose Dasgupta S, Ashok D, Pieters J, Tacchini-Cottier F, Rolink A, Acha-Orbea H, Finke D. Activated group 3 innate lymphoid cells promote T-cell-mediated immune responses. Proc Natl Acad Sci U S A 111: 12835–12840, 2014. 21. Cantrell VA, Owens SE, Chandler RL, Airey DC, Bradley KM, Smith JR, Southard-Smith EM. Interactions between Sox10 and EdnrB modulate penetrance and severity of aganglionosis in the Sox10Dom mouse model of Hirschsprung disease. Hum Mol Genet 13: 2289–2301, 2004. 22. Capoccia E, Cirillo C, Gigli S, Pesce M, D’Alessandro A, Cuomo R, Sarnelli G, Steardo L, Esposito G. Enteric glia: A new player in inflammatory bowel diseases. Int J Immunopathol Pharmacol 28: 443–451, 2015. 23. Cario E. Toll-like receptors in inflammatory bowel diseases: A decade later. Inflamm. Bowel Dis. 16 Oxford Academic: 1583–1597, 2010. 41 24. Cervi AL, Lukewich MK, Lomax AE. Neural regulation of gastrointestinal inflammation: role of the sympathetic nervous system. Auton Neurosci 182: 83–8, 2014. 25. Chastain EML, Duncan DS, Rodgers JM, Miller SD. The role of antigen presenting cells in multiple sclerosis. Biochim. Biophys. Acta - Mol. Basis Dis. 1812 NIH Public Access: 265–274, 2011. 26. Chatoor DR, Taylor SJ, Cohen CRG, Emmanuel A V. Faecal incontinence. Br J Surg 94: 134–144, 2007. 27. Cheadle G a, Costantini TW, Bansal V, Eliceiri BP, Coimbra R. Cholinergic Signaling in the Gut: A Novel Mechanism of Barrier Protection through Activation of Enteric Glia Cells. Surg Infect (Larchmt) 15: 387–93, 2014. 28. Chiu IM, Heesters BA, Ghasemlou N, Von Hehn CA, Zhao F, Tran J, Wainger B, Strominger A, Muralidharan S, Horswill AR, Bubeck Wardenburg J, Hwang SW, Carroll MC, Woolf CJ. Bacteria activate sensory neurons that modulate pain and inflammation. Nature 501: 52–7, 2013. 29. Cho JH. The genetics and immunopathogenesis of inflammatory bowel disease. Nat. Rev. Immunol. 8 Nat Rev Immunol: 458–466, 2008. 30. Chu H, Mazmanian SK. Innate immune recognition of the microbiota promotes host-microbial symbiosis. Nat. Immunol. 14 NIH Public Access: 668–675, 2013. 31. Cibrián D, Sánchez-Madrid F. CD69: from activation marker to metabolic gatekeeper. Eur. J. Immunol. 47 Wiley-VCH Verlag: 946–953, 2017. 32. Coelho-Aguiar J de M, Bon-Frauches AC, Gomes ALT, Verissimo CP, Aguiar DP, Matias D, Thomasi BB de M, Gomes AS, Brito GA de C, Moura-Neto V. The enteric glia: Identity and functions. Glia 63: 921–935, 2015. 33. Coombes JL, Powrie F. Dendritic cells in intestinal immune regulation. Nat. Rev. Immunol. 8 Europe PMC Funders: 435–446, 2008. 34. Crotzer VL, Blum JS. Autophagy and Its Role in MHC-Mediated Antigen Presentation. J Immunol 182: 3335–3341, 2009. 35. Cryan JF, O’riordan KJ, Cowan CSM, Sandhu K V., Bastiaanssen TFS, Boehme M, Codagnone MG, Cussotto S, Fulling C, Golubeva A V., Guzzetta KE, Jaggar M, Long-Smith CM, Lyte JM, Martin JA, Molinero-Perez A, Moloney G, Morelli E, Morillas E, O’connor R, Cruz-Pereira JS, Peterson VL, Rea K, Ritz NL, Sherwin E, Spichak S, Teichman EM, van de Wouw M, Ventura-Silva AP, Wallace-Fitzsimons SE, Hyland N, Clarke G, Dinan TG. The microbiota-gut-brain axis. Physiol Rev 99: 1877–2013, 2019. 36. Delvalle NM, Fried DE, Rivera-Lopez G, Gaudette L, Gulbransen BD. 42 Cholinergic activation of enteric glia is a physiological mechanism that contributes to the regulation of gastrointestinal motility. Am J Physiol - Gastrointest Liver Physiol 315: G473–G483, 2018. 37. Dengjel J, Schoor O, Fischer R, Reich M, Kraus M, Müller M, Kreymborg K, Altenberend F, Brandenburg J, Kalbacher H, Brock R, Driessen C, Rammensee H, Stevanovic S. Autophagy promotes MHC class II presentation of peptides from intracellular source proteins. Proc Natl Acad Sci U S A 102: 7922– 7, 2005. 38. Dewanjee S, Das S, Das AK, Bhattacharjee N, Dihingia A, Dua TK, Kalita J, Manna P. Molecular mechanism of diabetic neuropathy and its pharmacotherapeutic targets. Eur J Pharmacol 833: 472–523, 2018. 39. Dieleman LA, Ridwan BU, Tennyson GS, Beagley KW, Bucy RP, Elson CO. Dextran sulfate sodium-induced colitis occurs in severe combined immunodeficient mice. Gastroenterology 107: 1643–1652, 1994. 40. Dimitriadou V, Mécheri S, Koutsilieris M, Fraser W, Al-Daccak R, Mourad W. Expression of functional major histocompatility complex class II molecules on HMC-1 human mast cells. J Leukoc Biol 64: 791–799, 1998. 41. Dinarello CA. Proinflammatory cytokines. Chest 118: 503–508, 2000. 42. Du YT, Rayner CK, Jones KL, Talley NJ, Horowitz M. Gastrointestinal symptoms in diabetes: Prevalence, assessment, pathogenesis, and management. Diabetes Care 41: 627–637, 2018. 43. Escalante J, McQuade RM, Stojanovska V, Nurgali K. Impact of chemotherapy on gastrointestinal functions and the enteric nervous system. Maturitas 105 Elsevier Ireland Ltd: 23–29, 2017. 44. Flannagan RS, Jaumouillé V, Grinstein S. The cell biology of phagocytosis. Annu Rev Pathol Mech Dis 7: 61–98, 2012. 45. Fox CC, Jewell SD, Whitacre CC. Rat peritoneal mast cells present antigen to a PPD-specific T cell line. Cell Immunol 158: 253–264, 1994. 46. Freire MO, Van Dyke TE. Natural resolution of inflammation. Periodontol 2000 63: 149–164, 2013. 47. Friedrich M, Pohin M, Powrie F. Cytokine Networks in the Pathophysiology of Inflammatory Bowel Disease. Immunity 50: 992–1006, 2019. 48. Furness JB. The enteric nervous system and neurogastroenterology. Nat. Rev. Gastroenterol. Hepatol. 9: 286–294, 2012. 49. Furness JB, Costa M. Types of neurons in the enteric nervous system. J Auton 43 Nerv Syst 81: 87–96, 2000. 50. Gabella G. Ultrastructure of the nerve plexuses of the mammalian intestine: The enteric glial cells. Neuroscience 6: 425–436, 1981. 51. Gabella G, Trigg P. Size of neurons and glial cells in the enteric ganglia of mice, guinea-pigs, rabbits and sheep. J Neurocytol 13: 49–71, 1984. 52. Gallagher P, O’Mahony D. Constipation in old age. Best Pract Res Clin Gastroenterol 23: 875–887, 2009. 53. Geboes K, Rutgeerts P, Ectors N, Mebis J, Penninckx F, Vantrappen G, Desmet VJ. Major histocompatibility class II expression on the small intestinal nervous system in Crohn’s disease. Gastroenterology 103: 439–447, 1992. 54. Germain RN. t-cell development and the CD4-CD8 lineage decision. Nat Rev Immunol 2: 309–322, 2002. 55. Gershon MD, Rothman TP. Enteric glia. Glia 4: 195–204, 1991. 56. De Giorgio R, Guerrini S, Barbara G, Stanghellini V, De Ponti F, Corinaldesi R, Moses PL, Sharkey KA, Mawe GM. Inflammatory neuropathies of the enteric nervous system. Gastroenterology 126: 1872–1883, 2004. 57. De Giorgio R, Stanghellini V, Barbara G, Corinaldesi R, De Ponti F, Tonini M, Bassotti G, Sternini C. Primary enteric neuropathies underlying gastrointestinal motor dysfunction. Scand J Gastroenterol 35: 114–122, 2000. 58. Gomes P, Chevalier J, Boesmans W, Roosen L, Van Den Abbeel V, Neunlist M, Tack J, Vanden Berghe P. ATP-dependent paracrine communication between enteric neurons and glia in a primary cell culture derived from embryonic mice. Neurogastroenterol Motil 21, 2009. 59. Gong T, Liu L, Jiang W, Zhou R. DAMP-sensing receptors in sterile inflammation and inflammatory diseases. Nat. Rev. Immunol. 20 Nature Research: 95–112, 2020. 60. Greenwood-Van Meerveld B, Johnson AC, Grundy D. Gastrointestinal Physiology and Function. Handb Exp Pharmacol 239: 1–16, 2017. 61. Grider JR. Neurotransmitters mediating the intestinal peristaltic reflex in the mouse. J Pharmacol Exp Ther 307: 460–7, 2003. 62. Gulbransen BD, Bains JS, Sharkey KA. Enteric Glia Are Targets of the Sympathetic Innervation of the Myenteric Plexus in the Guinea Pig Distal Colon. J Neurosci 30: 6801–6809, 2010. 63. Gulbransen BD, Bashashati M, Hirota SA, Gui X, Roberts JA, MacDonald JA, 44 Muruve DA, McKay DM, Beck PL, Mawe GM, Thompson RJ, Sharkey KA. Activation of neuronal P2X7 receptor-pannexin-1 mediates death of enteric neurons during colitis. Nat Med 18: 600–604, 2012. 64. Gulbransen BD, Sharkey KA. Purinergic Neuron-to-Glia Signaling in the Enteric Nervous System. Gastroenterology 136: 1349–1358, 2009. 65. Gulbransen BD, Sharkey KA. Novel functional roles for enteric glia in the gastrointestinal tract. Nat. Rev. Gastroenterol. Hepatol. 9 Nature Publishing Group: 625–632, 2012. 66. Hansebout CR, Su CX, Reddy K, Zhang D, Jiang C, Rathbone MP, Jiang S. Enteric glia mediate neuronal outgrowth through release of neurotrophic factors. Neural Regen Res 7: 2165–2175, 2012. 67. Hato T, Dagher PC. How the innate immune system senses trouble and causes trouble. Clin J Am Soc Nephrol 10: 1459–1469, 2015. 68. Heller F, Fuss IJ, Nieuwenhuis EE, Blumberg RS, Strober W. Oxazolone colitis, a Th2 colitis model resembling ulcerative colitis, is mediated by IL-13- producing NK-T cells. Immunity 17: 629–638, 2002. 69. Hepworth MR, Monticelli LA, Fung TC, Ziegler CGK, Grunberg S, Sinha R, Mantegazza AR, Ma HL, Crawford A, Angelosanto JM, John Wherry E, Koni PA, Bushman FD, Elson CO, Eberl G, Artis D, Sonnenberg GF. Innate lymphoid cells regulate CD4 + T-cell responses to intestinal commensal bacteria. Nature 498: 113–117, 2013. 70. Hernandez S, Fried DE, Grubišić V, McClain JL, Gulbransen BD. Gastrointestinal neuroimmune disruption in a mouse model of Gulf War illness. FASEB J 33: 6168–6184, 2019. 71. Hernandez S, Morales-Soto W, Grubišić V, Fried D, Gulbransen BD. Pyridostigmine bromide exposure creates chronic, underlying neuroimmune disruption in the gastrointestinal tract and brain that alters responses to palmitoylethanolamide in a mouse model of Gulf War Illness. Neuropharmacology (2020). doi: 10.1016/j.neuropharm.2020.108264. 72. Herzog J, Maekawa Y, Cirrito TP, Illian BS, Unanue ER. Activated antigen- presenting cells select and present chemically modified peptides recognized by unique CD4 T cells. Proc Natl Acad Sci U S A 102: 7928–7933, 2005. 73. Hirst GD, McKirdy HC. Presynaptic inhibition at mammalian peripheral synapse? Nature 250: 430–1, 1974. 74. Hoff S, Zeller F, Von Weyhern CWH, Wegner M, Schemann M, Michel K, Rühl A. Quantitative assessment of glial cells in the human and guinea pig enteric nervous system with an anti-Sox8/9/10 antibody. J Comp Neurol 509: 356–371, 45 2008. 75. Holleran G, Lopetuso L, Petito V, Graziani C, Ianiro G, McNamara D, Gasbarrini A, Scaldaferri F. The innate and adaptive immune system as targets for biologic therapies in inflammatory bowel disease. Int. J. Mol. Sci. 18 MDPI AG: 2017. 76. Holvoet T, Devriese S, Castermans K, Boland S, Leysen D, Vandewynckel YP, Devisscher L, Van den Bossche L, Van Welden S, Dullaers M, Vandenbroucke RE, De Rycke R, Geboes K, Bourin A, Defert O, Hindryckx P, De Vos M, Laukens D. Treatment of Intestinal Fibrosis in Experimental Inflammatory Bowel Disease by the Pleiotropic Actions of a Local Rho Kinase Inhibitor. Gastroenterology 153: 1054–1067, 2017. 77. Hood L, Steinmetz M, Malissen B. Genes of the major histocompatibility complex of the mouse. Annu Rev Immunol 1: 529–68, 1983. 78. Hughes CE, Nibbs RJB. A guide to chemokines and their receptors. FEBS J. 285 Blackwell Publishing Ltd: 2944–2971, 2018. 79. 80. Iida T, Onodera K, Nakase H. Role of autophagy in the pathogenesis of inflammatory bowel disease. World J Gastroenterol 23: 1944–1953, 2017. Italiani P, Boraschi D. From monocytes to M1/M2 macrophages: Phenotypical vs. functional differentiation. Front. Immunol. 5 Frontiers Research Foundation: 2014. 81. Jabari S, De Oliveira EC, Brehmer A, Da Silveira ABM. Chagasic megacolon: Enteric neurons and related structures. Histochem. Cell Biol. 142 Springer Verlag: 235–244, 2014. 82. Jabbur SJ, El-Kak FH, Nassar CF. The enteric nervous system--an overview. Med Res Rev 8: 459–69, 1988. 83. Janeway CA, Medzhitov R. Innate immune recognition. Annu Rev Immunol 20: 197–216, 2002. 84. Jessen KR, Mirsky R. Astrocyte-like glia in the peripheral nervous system: an immunohistochemical study of enteric glia. J Neurosci 3: 2206–18, 1983. 85. Jones RS, Minogue AM, Connor TJ, Lynch MA. Amyloid-β-induced astrocytic phagocytosis is mediated by CD36, CD47 and RAGE. J Neuroimmune Pharmacol 8: 301–11, 2013. 86. Josefowicz SZ, Lu LF, Rudensky AY. Regulatory T cells: Mechanisms of differentiation and function. Annu. Rev. Immunol. 30 NIH Public Access: 531–564, 2012. 46 87. Kambayashi T, Laufer TM. Atypical MHC class II-expressing antigen-presenting cells: can anything replace a dendritic cell? Nat Rev Immunol 14: 719–30, 2014. 88. Kapur RP, Yost C, Palmiter RD. A transgenic model for studying development of the enteric nervous system in normal and aganglionic mice. [Online]. Development 116: 167–75, 1992. https://pubmed.ncbi.nlm.nih.gov/1483385/ [4 Oct. 2020]. 89. Keating DJ, Spencer NJ. Release of 5-hydroxytryptamine from the mucosa is not required for the generation or propagation of colonic migrating motor complexes. Gastroenterology 138: 659-70 670.e1–2, 2010. 90. Kermarrec L, Durand T, Neunlist M, Naveilhan P, Neveu I. Enteric glial cells have specific immunosuppressive properties. J Neuroimmunol 295–296: 79–83, 2016. 91. Kim BJ, Jones HP. Epinephrine-primed murine bone marrow-derived dendritic cells facilitate production of IL-17A and IL-4 but not IFN-γ by CD4+ T cells. Brain Behav Immun 24: 1126–1136, 2010. 92. Kimball BC, Mulholland MW. Enteric glia exhibit P2U receptors that increase cytosolic calcium by a phospholipase C-dependent mechanism. J Neurochem 66: 604–612, 1996. 93. King RHM. The role of glycation in the pathogenesis of diabetic polyneuropathy. J. Clin. Pathol. - Mol. Pathol. 54 BMJ Publishing Group: 400–408, 2001. 94. Koh TJ, DiPietro LA. Inflammation and wound healing: the role of the macrophage. Expert Rev. Mol. Med. 13 NIH Public Access: e23, 2011. 95. Kubinak JL, Round JL. Toll-like receptors promote mutually beneficial commensal-host interactions. PLoS Pathog 8: 1, 2012. 96. Kumar H, Kawai T, Akira S. Pathogen recognition by the innate immune system. Int Rev Immunol 30: 16–34, 2011. 97. Kunze WA, Furness JB. The enteric nervous system and regulation of intestinal motility. Annu Rev Physiol 61: 117–42, 1999. 98. Larabi A, Barnich N, Nguyen HTT. New insights into the interplay between autophagy, gut microbiota and inflammatory responses in IBD. Autophagy 16: 38– 51, 2020. 99. Latella G, Di Gregorio J, Flati V, Rieder F, Lawrance IC. Mechanisms of initiation and progression of intestinal fibrosis in IBD. Scand J Gastroenterol 50: 53–65, 2015. 100. Lee GR. The balance of th17 versus treg cells in autoimmunity. Int J Mol Sci 19: 47 1–14, 2018. 101. Levine B. Cell biology: Autophagy and cancer. Nature 446 Nature Publishing Group: 745–747, 2007. 102. Levine B, Kroemer G. Biological Functions of Autophagy Genes: A Disease Perspective. Cell 176 Cell Press: 11–42, 2019. 103. Liang XH, Kleeman LK, Jiang HH, Gordon G, Goldman JE, Berry G, Herman B, Levine B. Protection against Fatal Sindbis Virus Encephalitis by Beclin, a Novel Bcl-2-Interacting Protein. J Virol 72: 8586–8596, 1998. 104. Linden DR. Enhanced excitability of guinea pig ileum myenteric AH neurons during and following recovery from chemical colitis. Neurosci Lett 545: 91–95, 2013. 105. Linden DR, Sharkey KA, Mawe GM. Enhanced excitability of myenteric AH neurones in the inflamed guinea-pig distal colon. J. Physiol. 547 J Physiol: 589– 601, 2003. 106. Lomax AE, Sharkey KA, Furness JB. The participation of the sympathetic innervation of the gastrointestinal tract in disease states. Neurogastroenterol Motil 22: 7–18, 2010. 107. Lu Y, Li X, Liu S, Zhang Y, Zhang D. Toll-like receptors and inflammatory bowel disease. Front. Immunol. 9 Frontiers Media S.A.: 72, 2018. 108. Lundgren O, Svanvik J, Jivegård L. Enteric nervous system - I. Physiology and pathophysiology of the intestinal tract. Dig Dis Sci 34: 264–283, 1989. 109. Luo P, Liu D, Li C, He WX, Zhang CL, Chang MJ. Enteric glial cell activation protects enteric neurons from damage due to diabetes in part via the promotion of neurotrophic factor release. Neurogastroenterol Motil 30, 2018. 110. MacEachern SJ, Patel BA, Keenan CM, Dicay M, Chapman K, McCafferty D- M, Savidge TC, Beck PL, MacNaughton WK, Sharkey KA. Inhibiting Inducible Nitric Oxide Synthase in Enteric Glia Restores Electrogenic Ion Transport in Mice With Colitis. Gastroenterology 149: 445–55.e3, 2015. 111. Maceachern SJ, Patel BA, Mckay DM, Sharkey KA. Nitric oxide regulation of colonic epithelial ion transport: A novel role for enteric glia in the myenteric plexus. J Physiol 589: 3333–3348, 2011. 112. MacKenzie JR, Mattes J, Dent LA, Foster PS. Eosinophils Promote Allergic Disease of the Lung by Regulating CD4 + Th2 Lymphocyte Function . J Immunol 167: 3146–3155, 2001. 113. Mahdi BM. Role of HLA typing on Crohn’s disease pathogenesis. Ann Med Surg 48 4: 248–253, 2015. 114. Margolis KG, Gershon MD. Enteric Neuronal Regulation of Intestinal Inflammation. Trends Neurosci xx: 1–11, 2016. 115. Martinez-Vicente M, Cuervo AM. Autophagy and neurodegeneration: when the cleaning crew goes on strike. Lancet Neurol. 6 Lancet Neurol: 352–361, 2007. 116. Marwaha AK, Leung NJ, McMurchy AN, Levings MK. TH17 cells in autoimmunity and immunodeficiency : Protective orpathogenic? Front. Immunol. 3 Frontiers Media SA: 2012. 117. Mathew R, Karantza-Wadsworth V, White E. Role of autophagy in cancer. Nat. Rev. Cancer 7 Nat Rev Cancer: 961–967, 2007. 118. Matteoli G, Boeckxstaens GE. The vagal innervation of the gut and immune homeostasis. Gut 62: 1214–22, 2013. 119. Matteoli G, Gomez-Pinilla PJ, Nemethova A, Di Giovangiulio M, Cailotto C, van Bree SH, Michel K, Tracey KJ, Schemann M, Boesmans W, Vanden Berghe P, Boeckxstaens GE. A distinct vagal anti-inflammatory pathway modulates intestinal muscularis resident macrophages independent of the spleen. Gut (2013). doi: 10.1136/gutjnl-2013-304676. 120. Mckeown SJ, Stamp L, Hao MM, Young HM. Hirschsprung disease: A developmental disorder of the enteric nervous system. Wiley Interdiscip Rev Dev Biol 2: 113–129, 2013. 121. McQuade RM, Al Thaalibi M, Nurgali K. Impact of chemotherapy-induced enteric nervous system toxicity on gastrointestinal mucositis. Curr Opin Support Palliat Care 14: 293–300, 2020. 122. Medzhitov R. Origin and physiological roles of inflammation. Nature 454: 428– 435, 2008. 123. Middel P, Raddatz D, Gunawan B, Haller F, Radzun HJ. Increased number of mature dendritic cells in Crohn’s disease: Evidence for a chemokine mediated retention mechanism. Gut 55: 220–227, 2006. 124. Mittal M, Siddiqui MR, Tran K, Reddy SP, Malik AB. Reactive oxygen species in inflammation and tissue injury. Antioxidants Redox Signal. 20 Antioxid Redox Signal: 1126–1167, 2014. 125. Mizoguchi A. Animal models of inflammatory bowel disease. Prog Mol Biol Transl Sci 105: 263–320, 2012. 126. Mizoguchi A, Mizoguchi E. Animal models of IBD: linkage to human disease. Curr Opin Pharmacol 10: 578–87, 2010. 49 127. Mollaaghababa R, Pavan WJ. The importance of having your SOX on: Role of SOX10 in the development of neural crest-derived melanocytes and glia. Oncogene 22 Oncogene: 3024–3034, 2003. 128. Mourad FH, Saadé NE. Neural regulation of intestinal nutrient absorption. Prog Neurobiol 95: 149–162, 2011. 129. Moynes DM, Lucas GH, Beyak MJ, Lomax AE. Effects of inflammation on the innervation of the colon. Toxicol Pathol 42: 111–117, 2014. 130. Murakami M, Ohta T, Otsuguro KI, Ito S. Involvement of prostaglandin E2 derived from enteric glial cells in the action of bradykinin in cultured rat myenteric neurons. Neuroscience 145: 642–653, 2007. 131. Nakagawa I, Amano A, Mizushima N, Yamamoto A, Yamaguchi H, Kamimoto T, Nara A, Funao J, Nakata M, Tsuda K, Hamada S, Yoshimori T. Autophagy defends cells against invading group A Streptococcus. Science (80- ) 306: 1037– 1040, 2004. 132. Nasser Y, Ho W, Sharkey KA. Distribution of adrenergic receptors in the enteric nervous system of the guinea pig, mouse, and rat. J Comp Neurol 495: 529–553, 2006. 133. Nemeth ZH, Bogdanovski DA, Barratt-Stopper P, Paglinco SR, Antonioli L, Rolandelli RH. Crohn’s Disease and Ulcerative Colitis Show Unique Cytokine Profiles. Cureus 9, 2017. 134. Neunlist M, Aubert P, Toquet C, Oreshkova T, Barouk J, Lehur PA, Schemann M, Galmiche JP. Changes in chemical coding of myenteric neurones in ulcerative colitis. Gut 52: 84–90, 2003. 135. Neunlist M, Rolli-Derkinderen M, Latorre R, Van Landeghem L, Coron E, Derkinderen P, De Giorgio R. Enteric glial cells: Recent developments and future directions. Gastroenterology 147 W.B. Saunders: 1230–1237, 2014. 136. Neurath MF. Cytokines in inflammatory bowel disease. Nat Rev Immunol 14: 329–42, 2014. 137. Nickoloff BJ, Turka LA. Immunological functions of non-professional antigen- presenting cells: new insights from studies of T-cell interactions with keratinocytes. Immunol Today 15: 464–469, 1994. 138. Nijhuis LEJ, Olivier BJ, de Jonge WJ. Neurogenic regulation of dendritic cells in the intestine. Biochem Pharmacol 80: 2002–8, 2010. 139. Nishida A, Inoue R, Inatomi O, Bamba S, Naito Y, Andoh A. Gut microbiota in the pathogenesis of inflammatory bowel disease. Clin. J. Gastroenterol. 11 Springer Tokyo: 2018. 50 140. Nishino I. Autophagic Vacuolar Myopathy. Semin Pediatr Neurol 13: 90–95, 2006. 141. Oh K, Shen T, Le Gros G, Min B. Induction of Th2 type immunity in a mouse system reveals a novel immunoregulatory role of basophils. Blood 109: 2921– 2927, 2007. 142. Oishi Y, Manabe I. Macrophages in inflammation, repair and regeneration. Int Immunol 30: 511–528, 2018. 143. Oshima T, Miwa H. Gastrointestinal mucosal barrier function and diseases. J Gastroenterol 51: 768–778, 2016. 144. Panizzon CP do NB, Zanoni JN, Hermes-Uliana C, Trevizan AR, Sehaber CC, Pereira RVF, Linden DR, Neto MH de M. Desired and side effects of the supplementation with L-glutamine and L-glutathione in enteric glia of diabetic rats. Acta Histochem 118: 625–631, 2016. 145. Parkin J, Cohen B. An overview of the immune system. Lancet (London, England) 357: 1777–89, 2001. 146. Parzych KR, Klionsky DJ. An overview of autophagy: morphology, mechanism, and regulation. Antioxid Redox Signal 20: 460–73, 2014. 147. Perlmutter DH. The role of autophagy in α-1-antitrypsin deficiency: A specific cellular response in genetic diseases associated with aggregation-prone proteins. Autophagy 2 Taylor and Francis Inc.: 258–263, 2006. 148. Pesce M, Borrelli O, Saliakellis E, Thapar N. Gastrointestinal Neuropathies: New Insights and Emerging Therapies. Gastroenterol Clin North Am 47: 877–894, 2018. 149. Peterson LW, Artis D. Intestinal epithelial cells: Regulators of barrier function and immune homeostasis. Nat. Rev. Immunol. 14 Nature Publishing Group: 141– 153, 2014. 150. De Ponti F, Giaroni C, Cosentino M, Lecchini S, Frigo G. Adrenergic mechanisms in the control of gastrointestinal motility: from basic science to clinical applications. Pharmacol Ther 69: 59–78, 1996. Tramón P, Palomino P, Lahoz C, Leyva‐Cobian F. Eosinophil as antigen‐ 151. Pozo V Del, de Andrés B, Martín E, Cárdaba B, Fernández JC, Gallardo S, presenting cell: activation of T cell clones and T cell hybridoma by eosinophils after antigen processing. Eur J Immunol 22: 1919–1925, 1992. 152. Prame Kumar K, Nicholls AJ, Wong CHY. Partners in crime: neutrophils and monocytes/macrophages in inflammation and disease. Cell Tissue Res 371: 551– 565, 2018. 51 153. Qi R, Yang W, Chen J. Role of enteric glial cells in gastric motility in diabetic rats at different stages. J Huazhong Univ Sci Technol - Med Sci 33: 496–500, 2013. 154. Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F, Edberg S, Medzhitov R. Recognition of commensal microflora by toll-like receptors is required for intestinal homeostasis. Cell 118: 229–241, 2004. 155. Randhawa PK, Singh K, Singh N, Jaggi AS. A review on chemical-induced inflammatory bowel disease models in rodents. Korean J. Physiol. Pharmacol. 18 Korean Physiological Soc. and Korean Soc. of Pharmacology: 279–288, 2014. 156. Ravikumar B, Sarkar S, Davies JE, Futter M, Garcia-Arencibia M, Green- Thompson ZW, Jimenez-Sanchez M, Korolchuk VI, Lichtenberg M, Luo S, Massey DCO, Menzies FM, Moreau K, Narayanan U, Renna M, Siddiqi FH, Underwood BR, Winslow And AR, Rubinsztein DC. Regulation of mammalian autophagy in physiology and pathophysiology. Physiol. Rev. 90 American Physiological Society Bethesda, MD: 1383–1435, 2010. 157. van Rijt LS, Vos N, Hijdra D, de Vries VC, Hoogsteden HC, Lambrecht BN. Airway Eosinophils Accumulate in the Mediastinal Lymph Nodes but Lack Antigen-Presenting Potential for Naive T Cells. J Immunol 171: 3372–3378, 2003. 158. Roche PA, Furuta K. The ins and outs of MHC class II-mediated antigen processing and presentation. Nat Rev Immunol 15: 203–16, 2015. 159. Rock KL, Reits E, Neefjes J. Present Yourself! By MHC Class I and MHC Class II Molecules. Trends Immunol 37: 724–737, 2016. 160. Romagnani S. Th1/Th2 cells. Inflamm Bowel Dis 5: 285–94, 1999. 161. Romagnani S. T-cell subsets (Th1 versus Th2). Ann Allergy, Asthma Immunol 85: 9–18, 2000. 162. Rosenbaum C, Schick MA, Wollborn J, Heider A, Scholz C-J, Cecil A, Niesler B, Hirrlinger J, Walles H, Metzger M. Activation of Myenteric Glia during Acute Inflammation In Vitro and In Vivo. PLoS One 11: e0151335, 2016. 163. Rothman TP, Tennyson VM, Gershon MD. Colonization of the bowel by the precursors of enteric glia: Studies of normal and congenitally aganglionic mutant mice. J Comp Neurol 252: 493–506, 1986. 164. Rühl A. Glial cells in the gut. Neurogastroenterol Motil 17: 777–790, 2005. 165. Rühl A, Nasser Y, Sharkey KA. Enteric glia. Neurogastroenterol Motil 16 Suppl 1: 44–9, 2004. 166. Saffrey MJ. Cellular changes in the enteric nervous system during ageing. Dev Biol 382: 344–55, 2013. 52 167. Sakaguchi S, Yamaguchi T, Nomura T, Ono M. Regulatory T Cells and Immune Tolerance. Cell 133 Cell Press: 775–787, 2008. 168. Sanders KM, Koh SD, Ro S, Ward SM. Regulation of gastrointestinal motility- insights from smooth muscle biology. Nat. Rev. Gastroenterol. Hepatol. 9 Nat Rev Gastroenterol Hepatol: 633–645, 2012. 169. Santana MA, Esquivel-Guadarrama F. Cell biology of T cell activation and differentiation. Int Rev Cytol 250: 217–74, 2006. 170. Savidge TC, Newman P, Pothoulakis C, Ruhl A, Neunlist M, Bourreille A, Hurst R, Sofroniew M V. Enteric Glia Regulate Intestinal Barrier Function and Inflammation Via Release of S-Nitrosoglutathione. Gastroenterology 132: 1344– 1358, 2007. 171. Scheperjans F, Aho V, Pereira PAB, Koskinen K, Paulin L, Pekkonen E, Haapaniemi E, Kaakkola S, Eerola-Rautio J, Pohja M, Kinnunen E, Murros K, Auvinen P. Gut microbiota are related to Parkinson’s disease and clinical phenotype. Mov Disord 30: 350–358, 2015. 172. Schmid D, Pypaert M, Münz C. Antigen-loading compartments for major histocompatibility complex class II molecules continuously receive input from autophagosomes. Immunity 26: 79–92, 2007. 173. Schneider J, Jehle EC, Starlinger MJ, Neunlist M, Michel K, Hoppe S, Schemann M. Neurotransmitter coding of enteric neurones in the submucous plexus is changed in non-inflamed rectum of patients with Crohn’s disease. Neurogastroenterol Motil 13: 255–264, 2001. 174. Schulzke JD, Fromm M, Hegel U, Riecken EO. Ion transport and enteric nervous system (ENS) in rat rectal colon: mechanical stretch causes electrogenic Cl-secretion via plexus Meissner and amiloride-sensitive electrogenic Na- absorption is not affected by intramural neurons. Pflugers Arch 414: 216–21, 1989. 175. Segura BJ, Zhang W, Cowles RA, Xiao L, Lin TR, Logsdon C, Mulholland MW. Lysophosphatidic acid stimulates calcium transients in enteric glia. Neuroscience 123: 687–693, 2004. 176. Serhan CN, Levy BD. Resolvins in inflammation: Emergence of the pro-resolving superfamily of mediators. J. Clin. Invest. 128 American Society for Clinical Investigation: 2657–2669, 2018. 177. Sharma M, Hegde P, Aimanianda V, Beau R, Sénéchal H, Poncet P, Latgé JP, Kaveri S V., Bayry J. Circulating human basophils lack the features of professional antigen presenting cells. Sci Rep 3, 2013. 178. Shipkova M, Wieland E. Surface markers of lymphocyte activation and markers 53 of cell proliferation. Clin. Chim. Acta 413 Elsevier: 1338–1349, 2012. 179. Silva FAR, Rodrigues BL, Ayrizono MDLS, Leal RF. The Immunological Basis of Inflammatory Bowel Disease. Gastroenterol. Res. Pract. 2016 Hindawi Publishing Corporation: 2016. 180. da Silveira ABM, Lemos EM, Adad SJ, Correa-Oliveira R, Furness JB, D’Avila Reis D. Megacolon in Chagas disease: a study of inflammatory cells, enteric nerves, and glial cells. Hum Pathol 38: 1256–1264, 2007. 181. Soret R, Schneider S, Bernas G, Christophers B, Souchkova O, Charrier B, Righini-Grunder F, Aspirot A, Landry M, Kembel S, Faure C, Heuckeroth R, Pilon N. Glial Cell Derived Neurotrophic Factor Induces Enteric Neurogenesis and Improves Colon Structure and Function in Mouse Models of Hirschsprung Disease. Gastroenterology (2020). doi: 10.1053/j.gastro.2020.07.018. 182. Spencer NJ, Dinning PG, Brookes SJ, Costa M. Insights into the mechanisms underlying colonic motor patterns. J Physiol 594: 4099–116, 2016. 183. Sprent J. Antigen-Presenting Cells: Professionals and amateurs. Curr Biol 5: 1095–1097, 1995. 184. Steinmetz M, Hood LE. Genes of the major histocompatibility complex in mouse and man. Biotechnol Biol Front : 303–317, 2019. 185. Sternini C. Structural and chemical organization of the myenteric plexus. Annu Rev Physiol 50: 81–93, 1988. 186. Straub RH, Wiest R, Strauch UG, Härle P, Schölmerich J. The role of the sympathetic nervous system in intestinal inflammation. Gut 55: 1640–9, 2006. 187. Strawbridge AB, Blum JS. Autophagy in MHC class II antigen processing. Curr Opin Immunol 19: 87–92, 2007. 188. Strober W, Fuss IJ. Proinflammatory cytokines in the pathogenesis of inflammatory bowel diseases. Gastroenterology 140: 1756–1767, 2011. 189. Stuart PM. Major Histocompatibility Complex (MHC): Mouse. eLS : 1–7, 2015. 190. Su H, Na N, Zhang X, Zhao Y. The biological function and significance of CD74 in immune diseases. Inflamm Res 66: 209–216, 2017. 191. Sullivan PB. Hirschprung’s disease. Arch Dis Child 74: 5–7, 1996. 192. Takiishi T, Fenero CIM, Câmara NOS. Intestinal barrier and gut microbiota: Shaping our immune responses throughout life. Tissue barriers 5: e1373208, 2017. 54 193. Talamonti G, D’Aliberti G, Cenzato M. Aulus Cornelius Celsus and the Head Injuries. World Neurosurg. 133 Elsevier Inc.: 127–134, 2020. 194. Taylor GS, Bywater RA. Intrinsic control of the gut. Baillieres Clin Gastroenterol 2: 1–22, 1988. 195. Teitelbaum DH, Caniano DA, Qualman SJ. The pathophysiology of Hirschsprung’s-associated enterocolitis: importance of histologic correlates. J Pediatr Surg 24: 1271–1277, 1989. 196. Terman A, Brunk UT. Autophagy in cardiac myocyte homeostasis, aging, and pathology. Cardiovasc. Res. 68 Cardiovasc Res: 355–365, 2005. 197. Tesmer LA, Lundy SK, Sarkar S, Fox DA. Th17 cells in human disease. Immunol. Rev. 223 NIH Public Access: 87–113, 2008. 198. Timmermans JP, Hens J, Adriaensen D. Outer submucous plexus: an intrinsic nerve network involved in both secretory and motility processes in the intestine of large mammals and humans. Anat Rec 262: 71–8, 2001. 199. Trivedi PJ, Adams DH. Chemokines and chemokine receptors as therapeutic targets in inflammatory bowel disease; Pitfalls and promise. J. Crohn’s Colitis 12 Oxford University Press: S641–S652, 2018. 200. Tsuboi K, Nishitani M, Takakura A, Imai Y, Komatsu M, Kawashima H. Autophagy protects against colitis by the maintenance of normal gut microflora and secretion of mucus. J Biol Chem 290: 20511–20526, 2015. 201. Turco F, Sarnelli G, Cirillo C, Palumbo I, De Giorgi F, D’Alessandro A, Cammarota M, Giuliano M, Cuomo R. Enteroglial-derived S100B protein integrates bacteria-induced Toll-like receptor signalling in human enteric glial cells. Gut 63: 105–15, 2014. 202. Uranga-Ocio JA, Bastús-Díez S, Delkáder-Palacios D, García-Cristóbal N, Leal-García MÁ, Abalo-Delgado R. Enteric neuropathy associated to diabetes mellitus. [Online]. Rev Esp Enferm Dig 107: 366–73, 2015. http://www.ncbi.nlm.nih.gov/pubmed/26031865. 203. Uribe-Quero E, Rosales C. Control of phagocytosis by microbial pathogens. Front. Immunol. 8 Frontiers Media S.A.: 24, 2017. 204. De Vadder F, Grasset E, Holm LM, Karsenty G, Macpherson AJ, Olofsson LE, Bäckhed F. Gut microbiota regulates maturation of the adult enteric nervous system via enteric serotonin networks. Proc Natl Acad Sci U S A 115: 6458–6463, 2018. 205. Valečka J, Almeida CR, Su B, Pierre P, Gatti E. Autophagy and MHC-restricted antigen presentation. Mol Immunol 99: 163–170, 2018. 55 206. Vanner S, Surprenant A. Neural reflexes controlling intestinal microcirculation. Am J Physiol 271: G223-30, 1996. 207. Vasina V, Barbara G, Talamonti L, Stanghellini V, Corinaldesi R, Tonini M, De Ponti F, De Giorgio R. Enteric neuroplasticity evoked by inflammation. Auton Neurosci Basic Clin 126–127: 264–272, 2006. 208. Vera G, Castillo M, Cabezos PA, Chiarlone A, Martín MI, Gori A, Pasquinelli G, Barbara G, Stanghellini V, Corinaldesi R, De Giorgio R, Abalo R. Enteric neuropathy evoked by repeated cisplatin in the rat. Neurogastroenterol Motil 23: 370, 2011. 209. Verheijden S, De Schepper S, Boeckxstaens GE. Neuron-macrophage crosstalk in the intestine: a “microglia” perspective. Front Cell Neurosci 9: 403, 2015. 210. Ward MA, Pierre JF, Leal RF, Huang Y, Shogan B, Dalal SR, Weber CR, Leone VA, Musch MW, An GC, Rao MC, Rubin DT, Raffals LE, Antonopoulos DA, Sogin ML, Hyman NH, Alverdy JC, Chang EB. Insights into the pathogenesis of ulcerative colitis from a murine model of stasis-induced dysbiosis, colonic metaplasia, and genetic susceptibility. Am J Physiol - Gastrointest Liver Physiol 310: G973–G988, 2016. 211. Wardowska A. The epigenetic face of lupus: Focus on antigen-presenting cells. Int. Immunopharmacol. 81 Elsevier B.V.: 106262, 2020. 212. Wildenberg ME, Vos ACW, Wolfkamp SCS, Duijvestein M, Verhaar AP, Te Velde AA, Van Den Brink GR, Hommes DW. Autophagy attenuates the adaptive immune response by destabilizing the immunologic synapse. Gastroenterology 142: 1493-1503.e6, 2012. 213. Williams A, Jahreiss L, Sarkar S, Saiki S, Menzies FM, Ravikumar B, Rubinsztein DC. Aggregate-Prone Proteins Are Cleared from the Cytosol by Autophagy: Therapeutic Implications. Curr. Top. Dev. Biol. 76 Curr Top Dev Biol: 89–101, 2006. 214. Xue L, Farrugia G, Miller SM, Ferris CD, Snyder SH, Szurszewski JH. Carbon monoxide and nitric oxide as coneurotransmitters in the enteric nervous system: Evidence from genomic deletion of biosynthetic enzymes. Proc Natl Acad Sci U S A 97: 1851–1855, 2000. 215. Yamauchi T, Moroishi T. Hippo Pathway in Mammalian Adaptive Immune System. Cells 8: 398, 2019. 216. Yang H, Plevy SE, Taylor K, Tyan D, Fischel-Ghodsian N, McElree C, Targan SR, Rotter JI. Linkage of Crohn’s disease to the major histocompatibility complex region is detected by multiple non-parametric analyses. Gut 44: 519–526, 1999. 56 217. Yang L, Liu C, Zhao W, He C, Ding J, Dai R, Xu K, Xiao L, Luo L, Liu S, Li W, Meng H. Impaired autophagy in intestinal epithelial cells alters gut microbiota and host immune responses. Appl Environ Microbiol 84, 2018. 218. Yiu JHC, Dorweiler B, Woo CW. Interaction between gut microbiota and toll-like receptor: from immunity to metabolism. J. Mol. Med. 95 Springer Verlag: 13–20, 2017. 219. Yoo BB, Mazmanian SK. The Enteric Network: Interactions between the Immune and Nervous Systems of the Gut. Immunity 46: 910–926, 2017. 220. Yoshimoto T, Yasuda K, Tanaka H, Nakahira M, Imai Y, Fujimori Y, Nakanishi K. Basophils contribute to TH2-IgE responses in vivo via IL-4 production and presentation of peptide-MHC class II complexes to CD4+ T cells. Nat Immunol 10: 706–712, 2009. 221. Zhang C, Li L, Feng K, Fan D, Xue W, Lu J. ‘Repair’ Treg Cells in Tissue Injury. Cell Physiol Biochem 43: 2155–2169, 2017. 222. Zhang M, Sun K, Wu Y, Yang Y, Tso P, Wu Z. Interactions between Intestinal microbiota and host immune response in inflammatory bowel disease. Front Immunol 8: 1, 2017. 223. Zhang N, Weber A, Li B, Lyons R, Contag PR, Purchio AF, West DB. An inducible nitric oxide synthase-luciferase reporter system for in vivo testing of anti- inflammatory compounds in transgenic mice. J Immunol 170: 6307–6319, 2003. 224. Zhang W, Sarosi G, Barnhart D, Yule DI, Mulholland MW. Endothelin-activated calcium signaling in enteric glia derived from neonatal guinea pig. Am J Physiol - Gastrointest Liver Physiol 272, 1997. 225. Zhang YZ, Li YY. Inflammatory bowel disease: Pathogenesis. World J Gastroenterol 20: 91–99, 2014. 226. Zindel J, Kubes P. DAMPs, PAMPs, and LAMPs in Immunity and Sterile Inflammation. Annu. Rev. Pathol. Mech. Dis. 15 Annual Reviews Inc.: 493–518, 2020. 57 Potential roles of enteric glia in bridging neuroimmune communication in the gut CHAPTER 2: Work presented in this chapter has been published as: Chow AK, Gulbransen BD. Potential roles of enteric glia in bridging neuroimmune communication in the gut. Am J Physiol Gastrointest Liver Physiol. 2017 Feb 1;312(2):G145-G152. 58 Abstract The enteric nervous system (ENS) is a network of neurons and glia that controls ongoing gastrointestinal (GI) functions. Damage or injury to the ENS can lead to functional GI disorders. Current data support the conclusion that many functional GI disorders are caused by an imbalance between gut microbes and the immune system but how the ENS is involved in these interactions is less understood. Due to the proximity of the ENS to bacteria and other foreign antigens present in the GI tract, it is important to prevent the passage of these antigens through the GI epithelium. If any foreign compounds manage to pass through, an immune response is triggered to prevent injury to the ENS and underlying structures. However, careful modulation of the inflammatory response is required to allow for adequate elimination of foreign antigens while avoiding inappropriate over-activation of the immune system as in autoimmune disorders. Enteric neurons and glial cells are capable of performing these immunomodulatory functions to provide adequate protection to the ENS. Here, we review recent studies examining the interactions between the ENS and the immune system, specifically focusing on enteric glial cells and their ability to modulate inflammation in the ENS. 59 Introduction The enteric nervous system (ENS) is the largest division of the autonomic nervous system and it is housed entirely within the walls of the gastrointestinal (GI) tract. Neural reflexes mediated through the ENS are responsible for coordinating most ongoing activities of the GI tract such as patterns of motility, secretions and local blood flow. Like the brain, the ENS is entirely composed of neurons and glial cells that are housed within ganglia in two ganglionated plexuses. The neural circuitry in the submucosal plexus is responsible for coordinating gut secretions, absorption and local blood flow while the myenteric plexus is primarily responsible for coordinating contractions and relaxations of gut smooth muscle involved in patterns of gut motility. Proper orchestration of GI functions requires that both plexuses are intact and maintain healthy interactions with target cells. Injuries inflicted on the ENS during local inflammation disrupt the functional connectivity of enteric neural networks and create long–lasting changes in ENS function (13). These changes in the local control of gut reflexes contribute to a broad range of common functional GI disorders such as irritable bowel syndrome (IBS) and functional constipation and contribute to persistent intestinal dysmotility during remission in inflammatory bowel disease (IBD). Clearly, cells within the innate and adaptive arms of the immune system are the main drivers of gut inflammation. Generally speaking, inflammation is a protective response to invading pathogens that evade the first line of defense posed by the intestinal epithelial barrier and defects in the balanced immune response to intestinal bacteria is a major contributing factor to gut disease. The acute inflammatory response predominantly involves the recruitment and activation of the innate arm of the immune system (30) while 60 chronic inflammation involves both innate and adaptive components. Likewise, an overly aggressive adaptive immune response can attack native antigens in the gut and contribute to the development of inflammation (28). Current animal models used to study bowel inflammation are based on these principles and act to either increase epithelial permeability, such as the dextran sodium sulfate colitis model, or to enhance T-cell stimulation, such as the trinitrobenzene sulfonic acid (TNBS), dinitrobenzene sulfonic acid, oxazolone, T-cell adoptive transfer, and interleukin (IL)-10 knockout models (38). The understanding of the immune response in colitis is becoming increasingly refined and has led to many important breakthroughs in therapies. However, how inflammation produces long–lasting changes in gut function through effects on the ENS is still poorly understood. Importantly, new data suggest that bi-directional interactions between cells in the ENS and immune cells may actively participate in the modulation of inflammatory responses themselves. Here, we provide a short review of recent studies that highlight potential roles for the ENS in the regulation of gut inflammation and particularly focus on the key roles of enteric glial cells. Contribution of enteric neurons to inflammation Many immunomodulatory effects of the parasympathetic and sympathetic branches of the autonomic nervous system have emerged in recent years and the general picture is that vagal parasympathetic innervation is anti-inflammatory while α-adrenergic sympathetic innervation is pro-inflammatory (6, 10). However, far less is known about how intrinsic enteric neurons influence immune responses in the gut. The current evidence to date strongly suggests that enteric neurons have major immunomodulatory capabilities. The relative contributions of direct neuron-immune cell communication and 61 indirect communication via intermediary cells like enteric glia is still largely unknown. It is likely that many neuro-immune interactions at the ganglionic interface are mediated through enteric glia because enteric glia delineate the boundaries of enteric ganglia and largely surround enteric neuron cell bodies. However, neuronal processes extending outside the ganglia have a high potential for direct interactions with various immune cells. The capacity of neurons to influence immune responses through both direct and indirect pathways likely accounts for observation that enteric neuron number and density influences intestinal inflammation (48). Margolis et al. found that transgenic mice with a hyperplastic ENS and higher density of enteric neurons developed greater colonic inflammation when exposed to TNBS as compared to wildtype animals (48). The fact that this study used a transgenic animal model where high neuronal density is present prior to the administration of TNBS strongly suggests that neuronal hyperplasia is not merely a response to induced colonic inflammation but could, in fact, be a risk factor for the development of colitis. These findings are particularly interesting in light of the discovery that enteric glia kill a significant portion of neurons during acute inflammation (14). Given the pro-inflammatory effect of enteric neurons, it is tempting to speculate that glial-driven neurodegeneration functions to protect against the neuronal potentiation of inflammation but this concept is still unproven at this point. How higher numbers of enteric neurons translates to worsening inflammation is not immediately clear but likely occurs through the neuronal release of cytokines and neurotransmitters/neuromodulators. For example, enteric neurons from patients with Crohn’s disease demonstrate high expression of pro-inflammatory prostaglandin D2 (44). Enteric neurons are also capable of secreting leukocyte chemoattractive factors that 62 include tumor necrosis factor-α (TNF-α), IL-6, the monocyte chemoattractant protein-1 and possibly IL-8 when challenged with pathogenic bacteria (15, 60) (Fig.2.1). Interestingly, enteric neurons do not increase their secretion of these chemoattractive factors in response to Gram-positive bacteria despite the ability of both Gram-positive and Gram-negative bacteria to activate NF-κβ signal transduction pathways in neurons (15). This may be achieved through the effects of concurrent Wnt signaling within enteric neurons because the stimulation of the Wnt pathway in the presence of LPS confers an anti-inflammatory phenotype in enteric neurons in vitro (41) (Fig.2.1). These findings suggest that neurons are capable of differentiating between beneficial and harmful bacteria and participate in defensive responses to clear potentially pathogenic species. However, the inherent ability of neurons to recruit leukocytes could be potentially detrimental when additional neurons are present and may contribute to enteric ganglionitis. Although untested at this point, aberrant neuronal chemoattractant secretion under these conditions could be an important mechanism that contributes to a pro- inflammatory environment and a predisposition to gastrointestinal disease. 63 Figure 2.1: Overview of enteric neuron responses to select signals. Pathways highlighted in red denote pro-inflammatory pathways while those highlighted in blue lead to anti-inflammatory effects. Enteric neuron interactions with other cell types are shown with a white background. 64 In addition to their pro-inflammatory potential, enteric neurons may also contribute to anti-inflammatory effects via the synthesis and secretion of factors like netrin-1. Netrin-1 is typically known for its role in neuronal development but its expression is also induced during hypoxia-induced tissue damage (59) and the secretion of this molecule can induce angiogenesis, the activation of tissue stem cells and bind neutrophil cell surface receptors such as A2B adenosine receptors (3, 36). The main effect of netrin-1 on neutrophils is to inhibit their migration and accumulation and may induce an egress away from the source of netrin-1 (3). As such, enteric neurons could theoretically synthesize and secrete netrin- 1 as a response to inflammatory damage for neuronal survival and promote regeneration, and a loss in netrin-1 secretion could theoretically contribute to inappropriate leukocyte recruitment (4) (Fig.2.1). However, other major sources of netrin-1 in the GI system include the colonic epithelium; therefore further studies are required to elucidate the extent to which the ENS contributes to netrin-induced anti-inflammatory effects. Although activation of the Wnt pathway and secretion of prostaglandin D2 can induce inflammation, they can also inhibit the progression of inflammation. As described earlier, activation of the Wnt pathway with a concurrent LPS costimulatory signal on enteric neurons cause anti-inflammatory effects via the release of IL-10 (41). Neuronal prostaglandin D2 can be metabolized into 15-deoxy-Δ12-14-prostaglandin J2, an anti- inflammatory compound (44). The dual functioning of the Wnt pathway and prostaglandin release suggests that enteric neurons may be important for the selective activation and ‘fine-tuning’ of inflammatory responses in the ENS. In addition to cytokines and chemokines, neurotransmitters present in the ENS such as ATP, acetylcholine, noradrenaline, vasoactive intestinal peptide (VIP), neuropeptide Y, calcitonin gene- 65 related peptide, substance P, and serotonin can modulate the activities of intestinal leukocytes (1, 29, 34, 49) (Fig.2.1). These neurotransmitters yield both pro- and anti- inflammatory effects and likely have a major influence on inflammatory processes in the ENS. Margolis and Gershon (2016) recently reviewed the roles of several neurotransmitters in the regulation of intestinal inflammation and readers are referred to this excellent summary for further details (47). To summarize, the ability for immune cells to interact with neurotransmitters suggests a close relationship and frequent communication between enteric neurons and the immune system. In addition to acting directly on immune cells, these neurotransmitters can act on intestinal mucosa and alter epithelial barrier permeability. For example, in vitro experiments demonstrate that neuropeptide Y increases the epithelial barrier permeability and exposes the ENS to potential pathogens (17), whereas cholinergic and VIPergic signaling maintains barrier integrity and decreases permeability (18, 39, 55) (Fig.2.1). The control of intestinal epithelial barrier function and the exposure of the ENS to potential pathogens can modulate the inflammatory state of the ENS. This may represent an indirect means by which enteric neurons exert their influence on the immune system, further reinforcing the involvement of the ENS in immunomodulation. Glia in the healthy intestine Enteric glial cells are historically regarded as a network of cells that establish the homeostatic environment within enteric ganglia and function to provide both structural and nutritional support for enteric neurons. They are found primarily in both the submucosal and myenteric plexuses of the ENS, but may also be present outside the ENS, such as in the mucosal lamina propria. They are a heterogeneous group of cells 66 that can be subdivided into several types based on morphology and expression profiling (8, 56). These glial cell subtypes respond differently to purinergic signaling, suggesting different glial subtypes activate in varying situations. However, specific roles for the distinct glial populations have yet to be elucidated. Beyond structural and nutritional support, subsets of enteric glial cells also exert influence over other nearby cells of the GI tract and have local immunomodulatory effects. These functions largely resemble those of astrocytes in the central nervous system (CNS), and in this way, it is thought that certain groups of enteric glia fulfill astrocytic-like roles in the ENS. The relationship between enteric neurons and enteric glial cells begins early and new data show that glial cells are necessary for the proper maturation of neurons in the ENS(7). Glial cell guidance of neuronal development occurs through purinergic signaling between the two cell types and impaired signaling can lead to decreased enteric neuron density and complexity (7). Purinergic signaling between enteric glia and neurons continues into adulthood and plays an important role in neural circuits that coordinate GI motility (33, 50, 51). These exciting discoveries suggest that alterations to purinergic intercellular signaling between neurons and glia could contribute to gut dysfunction by impairing the development and maturation of enteric neurons or by altering neuronal activity. In agreement, impairing glial purinergic signaling in adult mice hinders coordinated contractions of the GI tract and slows colonic transit time (51). Mucosal enteric glial cells also form interactions with non-neuronal cells such as enterocytes and enteroendocrine cells that are important in the maintenance of GI homeostasis. Glial cells influence the development and maturation of enterocytes and these roles contribute to the maintenance of an intact GI epithelial barrier (52). Less is 67 known about mucosal glia-enteroendocrine interactions but specialized connections between the two cell types suggest that glia may have the potential to influence gastrointestinal hormone release (9). The fundamental roles of barrier regulation and hormone release in GI physiology suggest that defective interactions between enteric glia and enterocytes or enteroendocrine cells could contribute to GI inflammation and possibly even metabolic diseases such as diabetes and obesity (9, 65). Glial cells are activated by cytokines and immunomodulatory signals The ability of enteric glial cells to respond to immunomodulatory signals such as cytokines, bacteria and neurotransmitters in the extracellular milieu suggests that glia mediate a significant amount of the cross talk between the ENS and the immune system. For example, glial cells can detect local levels of the pro-inflammatory cytokine IL-1, IL- 4, and TNF-α (12) (Fig.2.2). Binding of these cytokines can elicit glial cell activation similar to reactive gliosis in the CNS and contribute to inflammation and GI dysfunction (61). Likewise, the activation of glial toll-like receptors 2 and 4 by LPS and other bacterial components drives pro-inflammatory pathways that are important in the protective response against bacteria that successfully bypass the gut epithelial barrier (58, 62). Eposito et al. demonstrated that LPS-activated glial cells occupy a crucial role in the creation of a robust inflammatory response in the local ENS environment and that the inhibition of the NF-κβ pathway in the glia can effectively ameliorate colonic inflammation (24). However, LPS-activated cultured human glia can also increase transcription of growth factors and several anti-inflammatory genes (42). This paradoxical elevation in the transcription of growth factors and anti-inflammatory genes with simultaneous expression of pro-inflammatory genes hints at a complex regulation of expression that likely involves 68 negative feedback pathways. Interestingly, Turco et al. showed that cultured human enteric glial cells are able to distinguish pathogenic from probiotic bacteria and modulate their expression of toll-like receptors accordingly (62). Similarly, Liddo et al. showed that exposing mixed cultures of rat enteric neurons and glia to a combination of LPS and Wnt3a induces the secretion of anti-inflammatory agents and inhibits NF-κβ activity in vitro (41). These findings suggest that the glial response to bacterial components in the ENS and the decision of whether to undertake pro- or anti-inflammatory pathways depends on the identity of the bacteria and which signaling molecules are being co- expressed. 69 Figure 2.2: Overview of enteric glial cell responses to select signals. Pathways highlighted with a red background are pro-inflammatory, while those highlighted in blue yield anti-inflammatory effects. Glial cell interactions with other cell types are shown with a white background. Note that glial response to certain signals such as ATP and LPS depend on the context by which the glial cell is stimulated and may yield varying responses. 70 Purines are a major component of the “inflammatory soup” generated during tissue injury and purines contribute to the pathophysiology of many inflammatory diseases of the intestine. Enteric glia are highly responsive to purines and express receptors for ATP and derivatives such as ADP and adenosine. The glial response to ADP through the activation of P2Y1 receptors is the most well characterized and is involved in the coordination of neural circuits and the promotion of neuronal maturation under normal conditions (7, 50, 51). However, glial P2Y1 receptors also play an important role in the glial response to neuronal danger cues released during inflammation and the activation of glia in this context causes neurodegeneration (14). The cause of the vastly different glial responses to P2Y1 receptor activation appears to primarily involve the influence of pro-inflammatory mediators, such as nitric oxide, on downstream effectors like connexin- 43 hemichannels (14). Therefore, the response of glia to purinergic agonists depends heavily on the context in which they are activated. How the activation of glia by other purines influences their response to inflammation is much less clear. For example, cultured human enteric glia express adenosine receptors that primarily couple to intracellular signal transduction pathways that utilize cAMP (42). Given the potent anti- inflammatory effect of adenosine in the gut, it is likely that adenosine has similar anti- inflammatory actions on glia but how the activation of glia by adenosine influences gut physiology and pathophysiology is currently unknown. Finally, enteric glial cells are innervated by both sympathetic and parasympathetic neurons of the autonomic nervous system (32, 63) (Fig.2.2). Stimulation of glial cells via parasympathetic vagal innervation confers an anti-inflammatory and neuroprotective effect, similar to the effects of parasympathetic vagal stimulation on intestinal 71 macrophages (49). Specifically, the activation of glial cholinergic receptors in vitro promotes increased epithelial barrier function and inhibits the transcription of the pro- inflammatory factor NF-κβ (18). These findings support previous data showing that the beneficial effects of vagal nerve stimulation on the intestinal epithelium following burn injuries is mediated through effects on enteric glia (22). Conversely, sympathetic- mediated glia activation occurs through purinergic (32) and noradrenergic (54) signaling. As noted above, the activation of glia by ATP may contribute to a pro-inflammatory phenotype but how the activation of glia by norepinephrine influences glia is not known. In summary, glia respond to a diverse array of immunomodulatory mediators that include pro-inflammatory cytokines, bacterial compounds, purines and transmitters from the autonomic nervous system. Activated glial cells exhibit some characteristics similar to astrocytes undergoing reactive astrogliosis in the CNS such as increased calcium influx and signaling and an increase in glial acidic fibrillary protein or S100β expression (12, 18, 42). However, glial activation is context specific and the outcome response of glia depends on integrative processing of all cues available at that particular time. Glia express and secrete cytokines and pro-inflammatory signals Mounting evidence in recent years suggests that glial cells are not idle bystanders during injury and inflammation in the ENS. What this evidence shows is that when activated, glia can contribute to a pro-inflammatory environment by secreting cytokines and other immunomodulatory signals. For example, one response of glia that are activated by inflammatory cues in the gut includes alterations to their morphology and expression and/or secretion of key proteins such as S100β and glial fibrillary acidic protein, similar to the process of reactive astrogliosis in the CNS (57). Increased secretion 72 of S100β by glia contributes to inflammation by activating receptors for advanced glycation end-products. This in turn leads to the production of nitric oxide by inducible nitric oxide synthase, the formation of reactive oxygen species, increased oxidative stress in the local environment and neuronal damage in ex vivo and in vitro studies (20, 23, 42, 46, 62). It is important to note that extracellular concentrations of S100β in the micromolar range are needed for pro-inflammatory effects and that basal S100β secretion in the nanomolar range yields neuroprotective effects (23) (Fig.2.2). Interestingly, despite an increase in local S100β during inflammation of the GI tract, Celikbilek et al. show a counterintuitive decrease in serum levels of S100β in patients with ulcerative colitis (16), possibly due to damaged enteric glial cells secreting reduced S100β as a result of exposure to prolonged inflammatory stress. Serum S100β levels over time in IBD may show fluctuations depending on glial cell viability and may prove useful in determining chronicity of inflammation. The stimuli for glial S100β release are incompletely understood but do include toll-like receptor agonists (62) and also likely includes purinergic signaling. Glial activation by purines during inflammation involves similar pathways as S100β such as inducible nitric oxide synthase and the release of ATP from activated glial cells (14). Glial S100β and ATP release both have neurotoxic effects on enteric neurons and these mediators are key drivers of ENS damage during acute inflammation in vivo (14, 23) (Fig.2.2). The glial secretion of cytokines and chemokines is of specific interest among the repertoire of compounds secreted by enteric glia because of their direct immunologic effects. Specifically, recent in vivo and in vitro studies demonstrate that glia secrete a number of cytokines and chemokines that include interferon gamma (IFN-γ), chemokine 73 ligand 20, TNF-α and prostaglandin D2 (26, 44, 58) (Fig.2.2). However, conflicting data both support and refute the concept that enteric glia secrete TNF-α. For example, Coquenlorge et al. showed that glial cell cultures are unable to secrete TNF-α even when stimulated with LPS (21). Based on this data, they concluded that TNF-α in the ENS originates from enteric neurons and not from glia. In contrast, Guedia et al. found that enteric glial cell lines are capable of secreting TNF-α when stimulated with LPS and that the effect is further enhanced when exposed to Tat protein derived from HIV (31). These conflicting results might suggest that multiple pathways can converge to induce inflammation and that there are various regulatory systems in place to restrict inappropriate inflammatory activation. Once activated, the glial secretion of cytokines and chemokines likely play an important role in the activation and recruitment of immune cells and modulates inflammation. Beyond IFN-γ, chemokine ligand 20, and TNF-α, numerous other pro-inflammatory cytokines and chemokines undergo transcriptional changes in activated enteric glia in culture. However, detectable protein secretion of these compounds has not been measured from activated glia (42, 58). Glial cells act as antigen presenting cells The many similarities between enteric glial cells and astrocytes hints that enteric glia, like astrocytes, may function as antigen presenting cells. New data show that enteric glia have phagocytic capabilities because glia transplanted from the ENS into the brain are capable of degrading pre-formed Aβ amyloid plaques in a similar manner as resident astrocytes (25, 64). In vivo and in vitro studies demonstrate that human enteric glial cells from both the myenteric and submucosal plexuses are also capable of expressing major histocompatibility complex II molecules, especially after exposure to bacteria or parasites 74 (5, 27, 62). In addition, enteric glia in patients with megacolon due to Chagas disease express the T cell co-stimulatory molecules cluster of differentiation (CD)80 and CD86 on their cell surface (5). Taken together, these data show that enteric glia have the ability to engulf and subsequently, present antigens to innate and adaptive immune cells and hints that enteric glial antigen presentation may play an important role at the neuro-immune interface. The role of glial antigen presentation in the gut is not currently understood but it could theoretically function to prime the adaptive immune system to bacteria in the ENS. By the same token, it would be possible for enteric glia to present neuronal debris to the adaptive immune system, thereby triggering enteric ganglionitis and an auto-reactive immune response against ENS neurons similar to what is seen in multiple sclerosis (45). Enteric glia exert immunosuppressive and anti-inflammatory effects In line with the thought that glial cells play a neuroprotective role to maintain homeostasis, there is increasing evidence that glial cells are capable of immunosuppression and anti-inflammatory actions. These anti-inflammatory effects are controlled, in part, by the release of soluble glial compounds. Of specific interest are glia- related peptides and lipids that include glial derived neurotrophic factor (GDNF), glial derived S-nitrosoglutathione (GSNO) and 15-deoxy-Δ12,14-prostaglandin J2 because these compounds have beneficial effects on intestinal epithelial permeability, inflammation and neuron survival (2, 19, 35, 65) (Fig.2.2). GDNF signals through RET and GFRα receptors present on innate immune cells and enterocytes of the gut to decrease expression of pro-inflammatory cytokines and help maintain and strengthen the intestinal epithelial barrier, respectively (35, 52, 65). Specifically, Ibiza et al. demonstrated via in vivo and in vitro mouse studies that ligands from the GDNF family act on type 3 75 innate lymphoid cells, leading to a release of anti-inflammatory IL-22 and increased expression of repair genes in the gut epithelium (35). In addition, RET and GFRα expression has been demonstrated in B lymphocytes, CD4+ and CD8+ T lymphocytes (53) but the direct effects of GDNF on these cells have yet to be described. Recent studies with cultured human enteric glial cells derived from the myenteric plexus suggest that GDNF contributes to the inhibition of proliferation by previously activated CD8- and CD8+ T lymphocytes. However, it seems that direct cell-to-cell interactions, as opposed to secreted compounds, play a larger role in preventing T lymphocyte proliferation (37). Furthermore, Kermarrec et al. showed that enteric glia from patients with Crohn’s disease exerted a greater immunosuppressant effect than control glia, suggesting that activated glial cells play an important role in immunomodulation. Similarly, von Boyen et al. describe increased activated mucosal glial cells expressing increased GFAP and GDNF in patients with Crohn’s disease. They further show that pro-inflammatory cytokines are an effective stimulus for GDNF secretion in vitro (11). Taken together, effects of GDNF contribute to an anti-inflammatory phenotype to reduce epithelial damage, prevent neuronal loss, suppress activated T lymphocytes and improve gut function, potentially as a negative feedback mechanism to limit the damage due to inflammation and pro-inflammatory cytokines (11, 37, 43) (Fig.2.2). Similar to the actions of GDNF, in vitro and in vivo studies show that GSNO acts to promote a robust permeability barrier in the intestinal epithelium. This is achieved by inducing increased expression of zonula occludens-1 and occludin protein to form more tight junctions in the epithelium (19, 40). In addition, GSNO provides anti-inflammatory effects via the inhibition of NF-κβ pathways that promote the expression of pro-inflammatory agents (40). Finally, secretion of 15-deoxy-Δ12,14-prostaglandin J2 76 from cultured rat myenteric glia elicits neuroprotective effects through the transcription factor NF-E2 related factor 2 pathway leading to neuronal synthesis of glutathione in vitro (2). Therefore, enteric glia can effectively modulate and control the extent of local inflammation and shift the local ENS environment back towards homeostasis. Conclusions Enteric glial cells have gained increasing attention because of their diverse emerging roles in the modulation of gut health and disease. Increasing evidence supports the idea of cross-talk between glial cells and many cells including enterocytes, neurons and immune cells. Specifically, activated glial cells play a substantial role in both the progression and inhibition of inflammation in the ENS. The seemingly contradictory actions may be due to the amount of secretion and extracellular concentration of glial products, as seen in S100β secretion, or the signaling pathway by which glial cells are activated, as demonstrated by cholinergic-induced versus cytokine-induced activation. Regulation of glial activation has not been well studied but may give insights into the specific contexts by which certain activation pathways are chosen to yield either pro- or anti-inflammatory effects in the ENS. Furthermore, various pro- and anti- inflammatory genes are transcribed in parallel, though many upregulated genes never reach translation and expression. This suggests potentially complex expression regulation and that enteric glial cells may self-regulate, to an extent, via negative feedback. The possible phagocytic ability of enteric glia, along with their capability to induce oxidative stress in their local environment and secretion of cytokines likens enteric glial cells to that of astrocytes and the innate immune system. Their ability to express major 77 histocompatibility complex II and T-cell costimulatory molecules suggests that glia play an important role in antigen presentation to T-cells. Further, the ability of enteric glial cells to modulate inflammation and immune activation shows that glia are an important mediator of neuro-immune interactions. Understanding the specific contribution of glia towards ENS injury and neuronal viability will offer important new insight into the pathogenesis of inflammatory and functional GI disorders. 78 REFERENCES 79 REFERENCES 1. Abad C, Cheung-Lau G, Coûté-Monvoisin A-C, Waschek JA. Vasoactive intestinal peptide-deficient mice exhibit reduced pathology in trinitrobenzene sulfonic acid-induced colitis. Neuroimmunomodulation 22: 203–12, 2015. 2. Abdo H, Mahé MM, Derkinderen P, Bach-Ngohou K, Neunlist M, Lardeux B. The omega-6 fatty acid derivative 15-deoxy-Δ12,14-prostaglandin J2 is involved in neuroprotection by enteric glial cells against oxidative stress. J Physiol 590: 2739–50, 2012. 3. Aherne CM, Collins CB, Eltzschig HK. Netrin-1 guides inflammatory cell migration to control mucosal immune responses during intestinal inflammation. Tissue barriers 1: e24957, 2013. 4. Aherne CM, Collins CB, Masterson JC, Tizzano M, Boyle T a, Westrich J a, Parnes J a, Furuta GT, Rivera-Nieves J, Eltzschig HK. Neuronal guidance molecule netrin-1 attenuates inflammatory cell trafficking during acute experimental colitis. Gut 61: 695–705, 2012. 5. Barcelos Morais Da Silveira A, De Oliveira EC, Neto SG, Luquetti AO, Toshio Fujiwara R, Correa Oliveira R, Brehmer A, Barcelos A, Oliveira EC De, Neto SG, Luquetti AO, Toshio R, Correa R, Brehmer A. Enteroglial cells act as antigen-presenting cells in chagasic megacolon. Hum Pathol 42: 522–532, 2011. 6. Bellinger DL, Lorton D. Autonomic regulation of cellular immune function. Auton Neurosci 182: 15–41, 2014. 7. Le Berre-Scoul C, Chevalier J, Oleynikova E, Cossais F, Talon S, Neunlist M, Boudin H. A novel enteric neuron-glia coculture system reveals the role of glia in neuronal development. J Physiol 1542: 33–36, 2016. 8. Boesmans W, Lasrado R, Vanden Berghe P, Pachnis V. Heterogeneity and phenotypic plasticity of glial cells in the mammalian enteric nervous system. Glia 63: 229–241, 2015. 9. Bohórquez D V., Samsa LA, Roholt A, Medicetty S, Chandra R, Liddle RA. An enteroendocrine cell - Enteric glia connection revealed by 3D electron microscopy. PLoS One 9, 2014. 10. Bonaz, Bruno; Sinniger, Valerie; Pellissier S. Anti-inflammatory properties of the vagus nerve: potential therapeuitc implications of vagus nerve stimulation. J Physiol 82: 1496–1514, 2011. 11. Von Boyen GBT, Steinkamp M, Geerling I, Reinshagen M, Schäfer KH, Adler G, Kirsch J. Proinflammatory cytokines induce neurotrophic factor expression in enteric glia: A key to the regulation of epithelial apoptosis in Crohn’s disease. 80 Inflamm Bowel Dis 12: 346–354, 2006. 12. von Boyen GBT, Steinkamp M, Reinshagen M, Schäfer K-H, Adler G, Kirsch J. Proinflammatory cytokines increase glial fibrillary acidic protein expression in enteric glia. Gut 53: 222–8, 2004. 13. Brierley SM, Linden DR. Neuroplasticity and dysfunction after gastrointestinal inflammation. Nat Rev Gastroenterol Hepatol 11: 611–27, 2014. 14. Brown IAM, McClain JL, Watson RE, Patel BA, Gulbransen BD. Enteric Glia Mediate Neuron Death in Colitis Through Purinergic Pathways That Require Connexin-43 and Nitric Oxide. C Cell Mol Gastroenterol Hepatol 2: 77–91, 2016. 15. Burgueño JF, Barba A, Eyre E, Romero C, Neunlist M, Fernández E. TLR2 and TLR9 modulate enteric nervous system inflammatory responses to lipopolysaccharide. J Neuroinflammation 13: 187, 2016. 16. Celikbilek A, Celikbilek M, Sabah S, Tanık N, Borekci E, Dogan S, Akin Y, Baldane S, Deniz K, Yilmaz N, Ozbakir O, Yucesoy M. The Serum S100B Level as a Biomarker of Enteroglial Activation in Patients with Ulcerative Colitis. Int J Inflam 2014: 986525, 2014. 17. Chandrasekharan B, Jeppsson S, Pienkowski S, Belsham DD, Sitaraman S V, Merlin D, Kokkotou E, Nusrat A, Tansey MG, Srinivasan S, Author C. Tumor necrosis factor (TNF)-neuropeptide Y (NPY) crosstalk regulates inflammation, epithelial barrier functions and colonic motility. Inflamm Bowel Dis 19: 2535–2546, 2013. 18. Cheadle G a, Costantini TW, Bansal V, Eliceiri BP, Coimbra R. Cholinergic Signaling in the Gut: A Novel Mechanism of Barrier Protection through Activation of Enteric Glia Cells. Surg Infect (Larchmt) 15: 387–93, 2014. 19. Cheadle GA, Costantini TW, Lopez N, Bansal V, Eliceiri BP, Coimbra R. Enteric Glia Cells Attenuate Cytomix-Induced Intestinal Epithelial Barrier Breakdown. PLoS One 8: 1–11, 2013. 20. Cirillo C, Sarnelli G, Turco F, Mango A, Grosso M, Aprea G, Masone S, Cuomo R. Proinflammatory stimuli activates human-derived enteroglial cells and induces autocrine nitric oxide production. Neurogastroenterol Motil 23, 2011. 21. Coquenlorge S, Duchalais E, Chevalier J, Cossais F, Rolli-Derkinderen M, Neunlist M. Modulation of lipopolysaccharide-induced neuronal response by activation of the enteric nervous system. J Neuroinflammation 11: 202, 2014. 22. Costantini TW, Bansal V, Krzyzaniak M, Putnam JG, Peterson CY, Loomis WH, Wolf P, Baird A, Eliceiri BP, Coimbra R. Vagal nerve stimulation protects against burn-induced intestinal injury through activation of enteric glia cells. Am J Physiol Gastrointest Liver Physiol 299: G1308–G1318, 2010. 81 23. Donato R, Sorci G, Riuzzi F, Arcuri C, Bianchi R, Brozzi F, Tubaro C, Giambanco I. S100B’s double life: Intracellular regulator and extracellular signal. Biochim Biophys Acta - Mol Cell Res 1793: 1008–1022, 2009. 24. Esposito G, Capoccia E, Turco F, Palumbo I, Lu J, Steardo A, Cuomo R, Sarnelli G, Steardo L. Palmitoylethanolamide improves colon inflammation through an enteric glia/toll like receptor 4-dependent PPAR-α activation. Gut (2013). doi: 10.1136/gutjnl-2013-305005. 25. Esposito G, Sarnelli G, Capoccia E, Cirillo C, Pesce M, Lu J, Calì G, Cuomo R, Steardo L. Autologous transplantation of intestine-isolated glia cells improves neuropathology and restores cognitive deficits in β amyloid-induced neurodegeneration. Sci Rep 6: 22605, 2016. 26. Fagbemi AO, Torrente F, Puleston J, Lakhoo K, James S, Murch SH. Enteric Neural Disruption in Necrotizing Enterocolitis Occurs in Association With Myenteric Glial Cell CCL20 Expression. J Pediatr Gastroenterol Nutr 57: 788– 793, 2013. 27. Geboes K, Rutgeerts P, Ectors N, Mebis J, Penninckx F, Vantrappen G, Desmet VJ. Major histocompatibility class II expression on the small intestinal nervous system in Crohn’s disease. Gastroenterology 103: 439–447, 1992. 28. Geremia A, Biancheri P, Allan P, Corazza GR, Di Sabatino A. Innate and adaptive immunity in inflammatory bowel disease. Autoimmun Rev 13: 3–10, 2014. 29. Di Giovangiulio M, Verheijden S, Bosmans G, Stakenborg N, Boeckxstaens GE, Matteoli G. The neuromodulation of the intestinal immune system and its relevance in inflammatory bowel disease. Front Immunol 6, 2015. 30. van der Gracht E, Zahner S, Kronenberg M, van der Gracht E, Zahner S, Kronenberg M, Zahner S, Kronenberg M. When Insult Is Added to Injury: Cross Talk between ILCs and Intestinal Epithelium in IBD. Mediators Inflamm 2016, 2016. 31. Guedia J, Brun P, Bhave S, Fitting S, Kang M, Dewey WL, Hauser KF, Akbarali HI. HIV-1 Tat exacerbates lipopolysaccharide-induced cytokine release via TLR4 signaling in the enteric nervous system. Sci Rep 6: 31203, 2016. 32. Gulbransen BD, Bains JS, Sharkey KA. Enteric Glia Are Targets of the Sympathetic Innervation of the Myenteric Plexus in the Guinea Pig Distal Colon. J Neurosci 30: 6801–6809, 2010. 33. Gulbransen BD, Sharkey KA. Purinergic Neuron-to-Glia Signaling in the Enteric Nervous System. Gastroenterology 136: 1349–1358, 2009. 34. Hassani H, Lucas G, Rozell B, Ernfors P. Attenuation of acute experimental 82 colitis by preventing NPY Y1 receptor signaling. Am J Physiol Gastrointest Liver Physiol 288: G550–G556, 2005. 35. Ibiza S, García-cassani B, Ribeiro H, Carvalho T, Almeida L, Marques R, Misic AM, Bartow-McKenney C, Larson DM, Pavan WJ, Eberl G, Grice EA, Veiga-Fernandes H, Eberi G, Grice EA, Veiga-Fernandes H, Eberl G, Grice EA, Veiga-Fernandes H. Glial-cell-derived neuroregulators control type 3 innate lymphoid cells and gut defence. Nature 535: 440–443, 2016. 36. Ke T, Wu Y, Li L, Liu Y, Yao X, Zhang J, Kong D, Li C. Netrin-1 Ameliorates Myocardial Infarction-Induced Myocardial Injury: Mechanisms of Action in Rats and Diabetic Mice. 797: 787–797, 2014. 37. Kermarrec L, Durand T, Neunlist M, Naveilhan P, Neveu I. Enteric glial cells have specific immunosuppressive properties. J Neuroimmunol 295–296: 79–83, 2016. 38. Kiesler P, Fuss IJ, Strober W. Experimental Models of Inflammatory Bowel Diseases. Cell Mol Gastroenterol Hepatol 1: 154–170, 2015. 39. Lesko S, Wessler I, Gäbel G, Petto C, Pfannkuche H. Cholinergic modulation of epithelial integrity in the proximal colon of pigs. Cells Tissues Organs 197: 411– 420, 2013. 40. Li Z, Zhang X, Zhou H, Liu W, Li J. Exogenous S-nitrosoglutathione attenuates inflammatory response and intestinal epithelial barrier injury in endotoxemic rats. J Trauma Acute Care Surg 80: 977–984, 2016. 41. Di Liddo R, Bertalot T, Schuster A, Schrenk S, Tasso A, Zanusso I, Conconi MT, Schäfer KH. Anti-inflammatory activity of Wnt signaling in enteric nervous system: in vitro preliminary evidences in rat primary cultures. J Neuroinflammation 12: 1–19, 2015. 42. Liñán-Rico A, Turco F, Ochoa-Cortes F, Harzman A, Needleman BJ, Arsenescu R, Abdel-Rasoul M, Fadda P, Grants I, Whitaker E, Cuomo R, Christofi FL. Molecular Signaling and Dysfunction of the Human Reactive Enteric Glial Cell Phenotype. Inflamm Bowel Dis 22: 1, 2016. 43. Liu GX, Yang YX, Yan J, Zhang T, Zou YP, Huang XL, Gan HT. Glial-derived neurotrophic factor reduces inflammation and improves delayed colonic transit in rat models of dextran sulfate sodium-induced colitis. Int Immunopharmacol 19: 145–152, 2014. 44. Le Loupp A-G, Bach-Ngohou K, Bourreille A, Boudin H, Rolli-Derkinderen M, Denis MG, Neunlist M, Masson D. Activation of the prostaglandin D2 metabolic pathway in Crohn’s disease: involvement of the enteric nervous system. BMC Gastroenterol 15: 112, 2015. 83 45. Lüssi F, Zipp F, Witsch E. Dendritic cells as therapeutic targets in neuroinflammation. Cell. Mol. Life Sci. (2016). doi: 10.1007/s00018-016-2170-9. 46. MacEachern SJ, Patel BA, Keenan CM, Dicay M, Chapman K, McCafferty D- M, Savidge TC, Beck PL, MacNaughton WK, Sharkey KA. Inhibiting Inducible Nitric Oxide Synthase in Enteric Glia Restores Electrogenic Ion Transport in Mice With Colitis. Gastroenterology 149: 445–55.e3, 2015. 47. Margolis KG, Gershon MD. Enteric Neuronal Regulation of Intestinal Inflammation. Trends Neurosci xx: 1–11, 2016. 48. Margolis KG, Stevanovic K, Karamooz N, Li ZS, Ahuja A, D’Autréaux F, Saurman V, Chalazonitis A, Gershon MD. Enteric neuronal density contributes to the severity of intestinal inflammation. Gastroenterology 141: 588–98, 598.e1– 2, 2011. 49. Matteoli G, Gomez-Pinilla PJ, Nemethova A, Di Giovangiulio M, Cailotto C, van Bree SH, Michel K, Tracey KJ, Schemann M, Boesmans W, Vanden Berghe P, Boeckxstaens GE. A distinct vagal anti-inflammatory pathway modulates intestinal muscularis resident macrophages independent of the spleen. Gut (2013). doi: 10.1136/gutjnl-2013-304676. 50. McClain JL, Fried DE, Gulbransen BD. Agonist-Evoked Ca2+ Signaling in Enteric Glia Drives Neural Programs That Regulate Intestinal Motility in Mice. C Cell Mol Gastroenterol Hepatol 1: 631–645, 2015. 51. McClain JL, Grubišić V, Fried D, Gomez-Suarez RA, Leinninger GM, Sévigny J, Parpura V, Gulbransen BD. Ca2+ responses in enteric glia are mediated by connexin-43 hemichannels and modulate colonic transit in mice. Gastroenterology 146: 497-507.e1, 2014. 52. Meir M, Flemming S, Burkard N, Bergauer L, Metzger M, Germer C-T, Schlegel N. Glial cell line-derived neurotrophic factor promotes barrier maturation and wound healing in intestinal epithelial cells in vitro. Am J Physiol - Gastrointest Liver Physiol 309: G613–G624, 2015. 53. Meir M, Flemming S, Burkard N, Wagner J, Germer C-T, Schlegel N. The glial cell-line derived neurotrophic factor: a novel regulator of intestinal barrier function in health and disease. Am J Physiol - Gastrointest Liver Physiol 310: G1118– G1123, 2016. 54. Nasser Y, Ho W, Sharkey KA. Distribution of adrenergic receptors in the enteric nervous system of the guinea pig, mouse, and rat. J Comp Neurol 495: 529–553, 2006. 55. Neunlist M, Toumi FF, Oreschkova T, Denis M, Leborgne J, Laboisse CL, Galmiche J-P, Jarry A. Human ENS regulates the intestinal epithelial barrier permeability and a tight junction-associated protein ZO-1 via VIPergic pathways. 84 Am J Physiol - Gastrointest Liver Physiol 285: G1028–G1036, 2003. 56. Rao M, Nelms BD, Dong L, Salinas-Rios V, Rutlin M, Gershon MD, Corfas G. Enteric glia express proteolipid protein 1 and are a transcriptionally unique population of glia in the mammalian nervous system. Glia 63: 2040–2057, 2015. 57. Robel S, Sontheimer H. Glia as drivers of abnormal neuronal activity. Nat Neurosci 19: 28–33, 2016. 58. Rosenbaum C, Schick MA, Wollborn J, Heider A, Scholz C-J, Cecil A, Niesler B, Hirrlinger J, Walles H, Metzger M. Activation of Myenteric Glia during Acute Inflammation In Vitro and In Vivo. PLoS One 11: e0151335, 2016. 59. Rosenberger P, Schwab JM, Mirakaj V, Masekowsky E, Mager A, Morote- Garcia JC, Unertl K, Eltzschig HK. Hypoxia-inducible factor-dependent induction of netrin-1 dampens inflammation caused by hypoxia. Nat Immunol 10: 195–202, 2009. 60. Rumio C, Besusso D, Arnaboldi F, Palazzo M, Selleri S, Gariboldi S, Akira S, Uematsu S, Bignami P, Ceriani V, Ménard S, Balsari A. Activation of smooth muscle and myenteric plexus cells of jejunum via Toll-like receptor 4. J Cell Physiol 208: 47–54, 2006. 61. Stoffels B, Hupa KJ, Snoek SA, Van Bree S, Stein K, Schwandt T, Vilz TO, Lysson M, Veer CVT, Kummer MP, Hornung V, Kalff JC, De Jonge WJ, Wehner S. Postoperative ileus involves interleukin-1 receptor signaling in enteric glia. Gastroenterology 146: 176-187.e1, 2014. 62. Turco F, Sarnelli G, Cirillo C, Palumbo I, De Giorgi F, D’Alessandro A, Cammarota M, Giuliano M, Cuomo R. Enteroglial-derived S100B protein integrates bacteria-induced Toll-like receptor signalling in human enteric glial cells. Gut 63: 105–15, 2014. 63. Uesaka T, Young HM, Pachnis V, Enomoto H. Development of the intrinsic and extrinsic innervation of the gut. Dev. Biol. (2016). doi: 10.1016/j.ydbio.2016.04.016. 64. Wyss-Coray T, Loike JD, Brionne TC, Lu E, Anankov R, Yan F, Silverstein SC, Husemann J. Adult mouse astrocytes degrade amyloid-beta in vitro and in situ. Nat Med 9: 453–457, 2003. 65. Xiao W, Wang W, Chen W, Sun L, Li X, Zhang C, Yang H. GDNF is Involved in the Barrier-Inducing Effect of Enteric Glial Cells on Intestinal Epithelial Cells Under Acute Ischemia Reperfusion Stimulation. Mol Neurobiol 50: 274–289, 2014. 85 CHAPTER 3: Enteric glial cells induce Th17 and Treg activation via autophagy-mediated MHC II expression 86 Abstract Background and Aims Enteric glial cells express type II major histocompatibility complex (MHC II) molecules in Crohn’s disease and Chagas disease, but it is unclear whether the expressed molecules are functional. We examined the capabilities of enteric glia to act as an antigen presenting cell in vivo and have immunomodulatory effects. Methods We used a tamoxifen-inducible glial-specific MHC II knockout (Sox10CreERT2;IABfl/fl) to isolate effects of enteric glial antigen presentation. We tested potential peptide sources to be loaded onto glial MHC II by measuring phagocytic activity and autophagy activation. We also determined enteric glial immunomodulatory capabilities by measuring T-lymphocyte activation and serum and colonic tissue cytokine levels. Results Enteric glia express MHC II molecules in response to low dose IFNγ and LPS exposure in the absence of intestinal tissue damage or inflammation. Glial MHC II expression increases activation in CD4+ T-lymphocytes, particularly in the Th17 and Treg subtypes, but not in Th1 or Th2 subtypes. Enteric glial MHC II does not influence serum or regional colonic tissue cytokine levels but may influence local cytokine levels. Autophagy activation is necessary, but insufficient to induce enteric glial MHC II expression. 87 Conclusions Enteric glial MHC II expression is induced by pro-inflammatory stimuli IFNγ and LPS and is dependent on autophagy activation. The expression of MHC II modulates T- lymphocyte activation, particularly in the Th17 and Treg subpopulations, but does not lead to changes in serum or colonic tissue cytokine levels. Keywords: enteric glial cell, autophagy, major histocompatibility complex type II, T- lymphocyte 88 Introduction Antigen presentation is a crucial process that maintains immune homeostasis by dictating T-lymphocyte activation at the transition between the innate and adaptive arms of the immune system. Dysfunction in antigen presentation can lead to the under- or over-activation of the immune system, or immune activation against inappropriate targets(8, 74). As a consequence, presenting the correct antigens and directing immune activity against the proper targets is of prime importance in the gastrointestinal (GI) tract where bacteria, viruses, parasites, and other foreign particles are constant challenges. Dysfunction in antigen presentation is a major contributing factor to immune dysregulation in diseases such as systemic lupus erythematosus, multiple sclerosis, and inflammatory bowel disease (IBD) (8, 48, 53, 74, 80). Professional antigen presenting cells are the primary cell types responsible for the process of antigen presentation and utilize major histocompatibility complex (MHC) II molecules to present specific peptides to T-lymphocytes. The identity of presented peptides activates specific T-lymphocytes, and therefore modulates immune activation. Since this process is sensitive to slight changes in peptides presented on MHC II molecules (4, 28), CD74 molecules are utilized to block peptide loading onto newly synthesized MHC II until their fusion with phagolysosomes (60). Although phagocytosis of extracellular foreign particles is the primary source of peptides for presentation MHC II molecules, autophagolysosomes can also contribute intracellular and ‘self’ peptides for loading onto MHC II molecules (65, 67). Peptide-loaded MHC II molecules are then targeted to the cell surface for presentation to T-lymphocytes. 89 In addition to professional antigen presenting cells expressing MHC II molecules, a set of ‘atypical’ MHC II-expressing cells, such as endothelial cells, epithelial cells, mast cells, and glial cells demonstrate variable levels of MHC II expression (35). Among these, enteric glia have gained considerable interest based on their ability to affect both T-lymphocyte activation and neuronal signaling (17, 24, 36). Enteric glial cells express MHC II molecules in an inducible manner, and share similar characteristics with antigen presenting cells by expressing T-lymphocyte costimulatory molecules CD80 and CD86 (3, 23, 70). Enteric glia are capable of interacting with T-lymphocytes when expressing neoantigens (15), and co-culture studies suggest that enteric glia modulate T- lymphocyte activation and regulate the activity of innate lymphoid type 3 cells (31, 36). Enteric glia also express both pro-inflammatory (20, 46, 62) and anti-inflammatory (1, 10, 31, 79) molecules in vitro, and may exhibit immune-modulating functions similar to those of related glial populations in the central nervous system (13). Although glial-T- lymphocyte interactions have been alluded to by in vitro studies, whether enteric glia affect play a significant role as antigen presenting cells in vivo is not known. The goal of this study was to test the hypothesis that glial antigen presentation modulates immune homeostasis in the intestine through interactions with CD4+ T- lymphocytes. We tested our hypothesis using a mild proinflammatory stimulus composed of IFNγ and LPS to perturb immune homeostasis without driving overt inflammation and tested the role of glial MHC II by generating mice with a conditional ablation of MHC II in enteric glia. The data show that enteric glia express MHC II in response to IFNγ and LPS and that glial MHC II expression is necessary for adequate CD4+ T-lymphocyte activation. Ablating glial antigen presentation impaired the 90 activation of T-lymphocyte subsets expressing IL-17 and FoxP3, suggesting that glial antigen presentation influences Th17 and Treg activation. Glial antigen presentation was independent of an increase in phagocytic activity and required the activation of autophagy. Taken together, these results show that glial autophagy triggered in response to proinflammatory stimuli induces glial antigen presentation, which functions to modulate the activation of T-lymphocyte subsets involved in tolerance. Animals Materials and Methods All protocols involving animals were approved by the Michigan State University Institutional Animal Care and Use Committee. Male and female C57BL/6 mice between 14 to 16-week-old were used unless stated otherwise. Animals were housed in ventilated cages with a 12-hour light-dark cycle, and access to food and water ad libitum. Mice with a tamoxifen-inducible conditional deletion of major histocompatibility complex II (MHC II) in enteric glia (Sox10CreERT2;IABfl/fl) were generated in-house by breeding Sox10CreERT2+/- mice ((42), gift from Dr. Vassilis Pachnis, The Francis Crick Institute, London, England) with floxed IAB mice (B6.129X1-H2-Ab1tm1Koni/J, RRID:IMSR_JAX:013181, The Jackson Laboratory, Bar Harbor, ME). RiboTag mice (Rpl22tm1.1Psam/J) expressing hemagglutinin on ribosomal protein L22 (Rpl22; 011029;RRID:IMSR_JAX:011029; The Jackson Laboratory, Bar Harbor, ME) were bred with the glial Sox10CreERT2 driver line to generate Sox10CreERT2+/-;Rpl22tm1.1Psam/J mice for experiments assessing glial transcriptional profiles as described previously (16). Enteric 91 glial reporter mice (Sox10CreERT2+/-;tdTomato) were generated by breeding Sox10CreERT2+/- mice with Ai14 mice (B6.Cg-Gt(ROSA)26Sortm14(CAG-tdTomato)Hze/J; RRID: IMSR_JAX:007914; The Jackson Laboratory, Bar Harbor, ME). CreERT2 activity was activated by feeding animals a 400mg/kg tamoxifen citrate diet for 2 weeks, and used for experiments immediately after the 2-week tamoxifen diet. Induction of glial MHC II Mice were injected with mouse recombinant interferon-gamma (IFNγ) (1µg/mouse, intraperitoneal (IP), cat# BMS326, eBioscience, Grand Island, NY), followed by an IP injection of E.coli O111:B4 lipopolysaccharide (LPS) (300µg/kg, cat# L4391, Millipore Sigma, St. Louis, MO) 2 hours afterwards to induce MHC II expression in enteric glia. Animals were scored for sickness severity and weights were measured at injection, and prior to sacrifice. Scoring for sickness severity was adapted from Shrum et al. (66). Briefly, animals were scored from 0-4 on each of the following categories: appearance, level of consciousness, activity, response to stimulus, eyes appearance, respiration rate, and respiration quality. Animals that had a sickness severity score of 21 or greater or lost >15% of body weight were euthanized and not included in analysis. Animals were euthanized and colons harvested at stated timepoints following LPS injection. Phagocytosis assay Phagocytic activity was assessed in longitudinal muscle-myenteric plexus (LMMP) whole mount preparations in vitro. Colons were isolated from transgenic mice expressing tdTomato in glia (Sox10CreERT2+/-;tdTomato) and whole mounts of myenteric plexus were prepared by removing the mucosa, submucosa, and circular muscle layers 92 by microdissection. Dissected tissues were incubated in Dulbecco’s Modified Eagle Medium (DMEM) containing IFNγ (10ng/mL, cat# BMS326, eBioscience, Grand Island, NY), LPS (5μg/mL, cat# L4391, Millipore Sigma, St. Louis, MO), and AlexaFluor488 conjugated mouse anti-CD45.2 antibody (cat#109816, BioLegend, San Diego, CA) and phagocytic substrates. Substrates were green pHrodo conjugated E. coli BioParticles (1mg/mL, cat# P35366, Thermo Fisher Scientific, Waltham, MA) AlexaFluor 488 labeled LPS (cat#L23351, Thermo Fisher Scientific, Waltham, MA) or FluoSpheres (1.0µm, cat#F8803, Thermo Fisher Scientific, Waltham, MA) for 2 hours at 37oC. pHrodo conjugated E. coli BioParticles fluoresce only in low pH environments such as within phagolysosomes. Fluorescence was visualized by confocal or epifluorescence and brightfield microscopy. Whole-Mount Immunofluorescence Colons were harvested from mice, fecal pellets flushed with phosphate buffered saline (PBS), and tissues fixed with Zamboni’s fixative at 4oC overnight. Fixed tissues were micro-dissected to remove the mucosa, submucosal plexus, and circular muscle layers. The remaining longitudinal muscle-myenteric plexus preparation were processed via immunofluorescence as described in prior work (24). Briefly, tissue preparations were washed three times at room temperature with Triton X-100 (0.1%, Sigma-Aldrich, St. Louis, MO) in PBS for 10 minutes each. Tissue were then incubated in blocking solution containing 4% normal goat serum, 0.4% Triton X-100, and 1% bovine serum albumin in PBS for 45 minutes at room temperature. Following blocking, samples were incubated with primary antibodies for 72 hours at 4oC. Tissues underwent three 10-minute washes in PBS prior to a 2-hour incubation with secondary antibodies at room temperature. 93 Following two 10-minute washes in PBS, labeled tissues were washed once in 0.1M phosphate buffer for 10 minutes and mounted onto microscope slides. Antibodies and dilutions used are listed in Table 3.1. Labeled tissues were visualized by epifluorescence microscopy with a 40X objective (0.75 numerical aperture; Plan Fluor, Nikon, Melville, NY) on a Nikon Eclipse Ni upright microscope (Melville, NY) using a QImaging Retiga 2000R camera (Teledyne Photometrics, Tucson, AZ), or by confocal microscopy using an Olympus Fluoview FV1000 microscope (Olympus, Center Valley, PA) with a 60x Plan-Apochromat oil immersion objective (1.42 numerical aperture). Immunofluorescence was quantified using Fiji software (https://imagej.nih.gov/ij/). Control experiments were performed prior to immunofluorescent labeling of samples by omitting primary antibodies in labeling protocols in order to control for non-specific binding and autofluorescence. H&E Staining Colon tissues were fixed overnight in Zamboni’s fixative at 4oC and washed with PBS. Tissues were embedded in paraffin and tissue sections (4-5µm thickness) were stained with hematoxylin and eosin by the MSU Investigative HistoPathology Laboratory. Frozen section Immunofluorescence Mesenteric lymph nodes and spleen were harvested from animals and fixed in Zamboni’s fixative at 4oC overnight. Fixed samples were washed with PBS and incubated in a 30% sucrose solution in 0.1M phosphate buffer at 4oC for 72 hours. Samples were submitted to the Michigan State University Histology Core for cryo- sectioning. Microscope slides with 6µm thick tissue sections were first warmed to room 94 temperature, rehydrated in distilled water, and warmed to 60oC. Following a 10-minute incubation in PBS at room temperature, tissue sections were labeled in a similar fashion as described above. Briefly, slides were incubated in PBS for 10 minutes at room temperature followed by two incubations of Triton X-100 (0.1%, Sigma-Aldrich, St. Louis, MO) in PBS for 10 minutes at room temperature. Slides were incubated with blocking solution containing 4% normal goat serum, 0.4% Triton X-100, and 1% bovine serum albumin in PBS for 45 minutes at room temperature. Following blocking, samples were incubated with primary antibodies for 72 hours at 4oC in a humidified slide chamber. Slides underwent three 10-minute washes in PBS prior to a 2-hour incubation with secondary antibodies at room temperature. Following two 10-minute washes in PBS, labeled slides were washed once in 0.1M phosphate buffer for 10 minutes, and mounted onto microscope slides. Antibodies and dilutions used are listed in Table 3.1. Flow cytometry Mouse mesenteric lymph nodes were harvested, and cells were isolated by mechanical dissociation and kept in FACS buffer consisting of PBS with 1% bovine serum albumin. Cells were passed through a 40μm cell strainer (Corning Life Sciences, Tewksbury MA) and centrifuged at 350g for 5 minutes at 4oC. Supernatant was removed and the cell pellet is washed once and resuspended in 1mL of ice-cold FACS buffer. To label cell surface markers, cells were incubated with conjugated antibodies for 30min at 4oC in the dark. Labeled cells were washed once more in FACS buffer before analysis with a BD LSR II flow cytometer (Becton, Dickinson and Company, Franklin Lakes, NJ). To label intracellular targets for flow cytometric analysis, mesenteric lymph node cells are first processed for cell surface labelling. Surface-labeled cells were fixed and 95 permeabilized with FoxP3 Fixation / Permeabilization buffer (eBioscience 00-5523-00, Grand Island, NY) overnight at 4oC in the dark. Fixed cells were washed with permeabilization buffer prior to incubation with intracellular-specific antibodies for 30min at 4oC in the dark. Cells were washed once more with permeabilization buffer prior to resuspension in ice-cold FACS buffer and analyzed with a BD LSR II flow cytometer (Becton, Dickinson and Company, Franklin Lakes, NJ). Flow cytometry data was analyzed with FCS Express 7 Research Edition (De Novo Software, Pasadena, CA). Antibodies and dilutions used are listed in Table 3.1. We eliminated doublets from flow cytometric analysis by plotting cells of interest on a forward scatter area vs height plot and gating for singlets. Resultant cells were plotted on a side scatter width vs height plot to ensure elimination of doublets. To properly control flow cytometric analysis of mesenteric lymph nodes, we incorporated unstained samples consisting of mesenteric lymph node cells incubated with FACS buffer without labeled antibodies to determine autofluorescence of mesenteric lymph node cells in each channel. Single stained controls consisted of UltraComp eBeads (Thermo Fisher Scientific, Waltham, WA) incubated with a single labeled antibody of each antigen of interest to calculate fluorescent compensation between each fluorophore. To identify the boundary between positive and negative fluorescence for each antigen of interest, we incorporated a panel of fluorescence minus one controls by labeling mesenteric lymph node cells with all fluorophores except the one being measured. Gene expression Sox10-RiboTag mice were used to quantify gene expression levels in enteric glial cells. Mice were treated with a 400mg/kg tamoxifen citrate diet for 2 weeks to induce glial- 96 specific ribosomal HA-tag expression. Colons were harvested, fecal pellets flushed with PBS, and tissue snap frozen on dry ice and stored at -80oC. The RNA extraction protocol was performed as described by Delvalle et al. with minor modifications (16). Briefly, homogenization buffer was made by mixing 7.38mL RNAse-free water with 1mL Nonident P-40 (cat#: 21-3277, Millipore Sigma, St. Louis, MO), 1mL of 1M potassium chloride, 500µL of pH 7.4 Tris 1.0, 120µL of 1M magnesium chloride, and 100mg of sodium deoxycholate. Frozen colon tissues were placed in 1mL of ice-cold supplemented homogenization buffer containing 949μL homogenization buffer, 40µL cycloheximide (Millipore Sigma, St. Louis, MO), 40µL protease inhibitor (cat#: P8340, Millipore Sigma, St. Louis, MO), 20µL RNAse inhibitor (cat#: AM2684, Thermo Fisher Scientific, Waltham, WA), 2µL dithiothreitol (cat#: 646563, Millipore Sigma, St. Louis, MO), and 3mg/mL heparin (cat#: H3149, Millipore Sigma, St. Louis, MO). Frozen tissues were homogenized using a GentleMACS Dissociator (Miltenyi Biotech, Auburn, CA) in M-tubes running the program RNA_02. Tissue homogenates were centrifuged at 10,000g at 4oC for 10min, supernatants collected, and incubated with 5μL of monoclonal anti-hemagglutinin (HA) antibody (16B12, BioLegend, San Diego, CA) at 4oC for 4 hours on a tube rotator. During antibody incubation, magnetic protein A/G beads (Thermo Fisher Scientific, Waltham, WA) were washed twice in a magnet with 500μL homogenization buffer. HA-labeled tissue homogenate supernatants were then incubated with the washed magnetic beads on a tube rotator at 4oC overnight. Following incubation, samples were washed three times in a magnet with 800μL of high-salt buffer. 350μL of RLT lysis buffer (Qiagen, Germantown, MD) were added to samples, and RNA released from magnetic beads by vortexing for 30 seconds. RNA was 97 collected by placing sample on a magnet and collecting the supernatant. Total RNA was purified using a RNeasy Micro kit (Qiagen, Germantown, MD) according to manufacturer’s instructions. Purified RNA was flash frozen and submitted to the UCLA Center for Systems Biomedicine core facility (UCLA, Los Angeles, CA) for analysis via a custom NanoString nCounter multiplex panel (NanoString, Seattle, WA). Gene expression levels were quantified by based on counts of fluorescent barcodes specific to individual genes. Cytokine quantification Serum and homogenized colon tissue samples were processed for cytokine quantification as described in prior work by Hernandez et al. with minor modifications (27). Briefly, serum samples were prepared by first harvesting whole blood from mice by cardiac puncture and allowed to clot by incubating at room temperature undisturbed for 15min in a covered test tube. Tubes were then centrifuged at 1500g at 4oC for 10 minutes. Serum was collected from the supernatant, diluted 2-fold with PBS to a final volume of 80µL, frozen at -80oC, and submitted to Eve Technologies (Calgary, AB, Canada) on dry ice for analysis. For tissue cytokine analysis, colon tissues were collected, and fecal pellets flushed prior to snap freezing on dry ice and storage at - 80oC. Frozen tissue samples are weighed and placed in 10% w/v of tris-buffered saline containing 0.05% tween-20, 25mM Tris-HCl, 130mM sodium chloride, 2.7mM potassium chloride, and SIGMAFAST protease inhibitor cocktail (1 tablet, cat#: S8820, Millipore Sigma, St. Louis, MO). Tissues are homogenized using a GentleMACS Dissociator (Miltenyi Biotech, Auburn, CA) in M tubes running the program Protein_01. Tissue homogenates were centrifuged at 4,000g at 4oC for 5min, and supernatants collected. 98 Supernatants were diluted with homogenization buffer to a protein concentration of 4mg/mL in a final volume of 75μL. Samples were frozen at -80oC prior to submission to Eve Technologies (Calgary, AB, Canada) for analysis via the mouse 31-plex multiplex cytokine array. Autophagy induction and detection Live whole mount preparations of myenteric plexus were prepared as described above and in prior work (24). Autophagy was induced by incubating samples with DMEM containing Rapamycin (50nM,cat#13346, Cayman Chemical, Ann Arbor, MI) for 16 hours at 37oC. Autophagy was inhibited by incubating samples with DMEM containing 3-MA (5mM, cat#13242, Cayman Chemical, Ann Arbor, MI) for 16 hours at 37oC. Autophagy was detected in enteric neurons and glia by measuring P62 immunofluorescence and LC3B puncta per cellular area. LC3B puncta was detected using FIJI by first thresholding the LC3B channel using the auto setting of the Intermodes algorithm in the ‘Threshold’ function of FIJI. LC3B puncta in enteric glia and neurons was measured in FIJI using the ‘Analyze Particles’ function, setting particle size of interest from 0 to Infinity and circularity from 0 to 1. Data Analysis Confocal immunofluorescent z-stack images were imported to FIJI software (https://imagej.nih.gove/ij/) as a Hyperstack using the Bio-Formats plugin (Open Microscopy Environment, University of Dundee, UK). To measure fluorescent intensity of a specific antigen within enteric glia or neurons, we first created a region of interest outlining GFAP-positive cells in each ganglion to isolate enteric glial expression. The 99 GFAP-positive region of interest was constructed in FIJI using the auto setting of the Otsu algorithm in the ‘Threshold’ function of FIJI. The resultant selection was converted into a region of interest in FIJI using the ‘Create Selection’ function. Total ganglionic expression of a given labeled antigen was measured by creating a ganglionic region of interest by utilizing the GFAP channel to outline the total ganglionic area. Glial fluorescent intensity or particle number was subtracted from total fluorescent intensity or particle number within the ganglion to calculate levels within enteric neurons. To calculate Mander’s overlap coefficient between two fluorophores, we utilized the FIJI plugin ‘Just Another Colocalisation Plugin’ (6), and adjusted thresholding parameters to include GFAP+ glial processes without incorporating background noise, or MHC II and CD74 signal without incorporating background noise. Additionally, using Metamorph software, we performed line-scans across enteric ganglion confocal z-projection images. Confocal z-stack images were processed by first converting z-stacks into maximum projection images. Background subtraction was applied in each channel to remove noise. Maximum gray values of labeled antigens were measured along the length of a line drawn within enteric ganglia to visualize co-presence of fluorophores. We used the 3D Viewer plugin in FIJI to create a 3-dimensional reconstruction of GFAP- positive ganglia and MHC II expression. 3-dimensional reconstruction is displayed as a volume with a resampling factor of 2. We analyzed flow cytometric data using FCS Express 7 Research Edition (De Novo Software, Pasadena, CA) and determined gating cutoffs for CD4, CD69, IL4, IL17, IFNγ, and FoxP3 markers using compensation and fluorescence minus one controls as described above. We determined the activated fraction of each T-lymphocyte subtype by calculating the proportion of T-lymphocyte 100 subtype marker and CD69 double positive cells within the total number of subtype marker positive cells. We used IFNγ as a marker for Th1, IL4 for Th2, IL17 for Th17, and FoxP3 for Treg subtypes. Nanostring gene expression data was processed by nSolver v4.0 software (NanoString Technologies, Seattle, WA). Data was curated to remove entries flagged by nSolver quality control or entries that have fewer reads than negative controls. Data is represented as fold change compared to the mean number of reads in saline control condition. For cytokine quantification, a four-parameter logistic regression was utilized to calculate the concentration of each analyte, and samples below or above the logistic regression were designated as 0 or the highest standard curve value, respectively. Statistics All data are represented as mean ± SEM and were analyzed by GraphPad Prism 7 (GraphPad Software, San Diego, CA). Immunofluorescence, flow cytometry, and quantified cytokine data were compared between genotype and treatment conditions using unpaired two-way ANOVA with Tukey-corrected multiple comparisons. Gene expression data retrieved from RNAseq databases are expressed as transcripts per million (tpm) and were compared by one-way ANOVA with Tukey-corrected multiple comparisons. A level of P < .05 was set as the cutoff for statistical significance. P values are denoted as follows: < .05 (*), < .005 (**), < .0005 (***), <.0001 (****). Outliers were detected in GraphPad Prism 7 using the ROUT method with Q=1%. 101 Antigen Target GFAP Host Species Dilution Conjugate Manufacturer Product no. Chicken 1:1000 n/a (primary Ab) Abcam ab4674 MHC II Rat CD74 P62 LC3B Anti-chicken IgY Rabbit Rabbit Rabbit Goat 1:200 1:200 1:200 1:200 1:400 n/a (primary Ab) Novus NBP2-21789 n/a (primary Ab) Bioss BS-2518R n/a (primary Ab) Abcam ab109012 n/a (primary Ab) Abcam ab192890 Dylight405 Jackson 103-475-155 Anti-rat IgG Goat 1:400 AlexaFluor 594 Jackson 112-585-003 Anti-rabbit IgG Goat 1:400 AlexaFluor 488 Invitrogen A-11034 CD45.2 Mouse 1:200 AlexaFluor488 BioLegend 109816 Table 3.1: Antibodies used in Chapter 3. 102 Table 3.1 (cont’d) CD4 CD8 CD69 IFNg IL-4 IL-17 FoxP3 Rat Rat Armenian Hamster Rat Rat Rat Rat 1:200 1:200 1:200 1:100 1:100 1:20 FITC BioLegend 100509 APC/Cy7 BioLegend 100714 PE/Cy7 BioLegend 104512 PE APC BioLegend 505808 BioLegend 504105 Brilliant Violet 510 BioLegend 506933 1:100 Pacific Blue BioLegend 126410 103 Results Conditional ablation of glial MHC II in Sox10CreERT2;IABfl/fl mice and IFNγ + LPS challenge Enteric glia express MHC II in response to proinflammatory stimuli such as LPS and IFNγ (Fig.3.1a, d-f), similar to findings by Turco et al. (70). These stimuli also elicit antigen presentation by professional immune cells which can mask the contribution of glia. Therefore, we used a minimal pro-inflammatory stimulus (IFNγ 1µg / mouse; LPS 0.3mg/kg; i.p.) to study glial effects in vivo without an overwhelming inflammatory response from other immune cells (Fig.3.1c). This challenge is sufficient to induce glial MHC II (Fig.3.1d-f) and causes minor weight loss (Fig.3.2a) but does not increase sickness behavior (Fig.3.2b), alter tissue architecture (Fig.3.2c), or cause significant histological damage (Fig.3.2d). Murine MHC II molecules are composed of gene products from the I-A or I-E loci. C57BL/6 mice have a H-2b MHC haplotype due to a deficient I-E loci and rely solely on the I-A loci for MHC II expression (29, 30, 39). We isolated the specific role of enteric glia by creating a tamoxifen-inducible, glial-specific MHC II knockout (KO) mouse model (Sox10CreERT2;IABfl/fl) where the IAB gene, a crucial component in the MHC II I-A loci, is deleted in Sox10+ enteric glia (Fig.3.1b,c). 104 Figure 3.1: Enteric glial cells express MHC II when stimulated with IFNγ and LPS. (A) Schematic depicting enteric glial expressing MHC II molecules as a response to exposure to 1µg of IFNγ and 0.3mg/kg LPS. (B) Schematic depicting gene targeting strategy in Sox10CreERT2;IABfl/fl mice. Tamoxifen-sensitive CreERT2 expression is confined to glia by the Sox10 promoter and excises the floxed H2-Ab1 exon 1 loci. (C) Schematic depicting exposure protocol. Wildtype and MHC II KO mice are fed 105 Figure 3.1 (cont’d): 400mg/kg tamoxifen in chow for 14 days to induce Cre recombinase and MHC II ablation. Animals are injected via IP with pro-inflammatory stimuli consisting of 1µg of IFNγ followed by 0.3mg/kg LPS 2 hours afterwards or 1µg of IFNγ and volume-matched saline as controls. Tissues were harvested 16 hours post-injection. (D) Representative confocal images of GFAP (grey) and MHC II (red) immunofluorescence labeling in enteric ganglia. Enteric glial cells express MHC II when stimulated with IFNγ and LPS. Muscularis macrophages (Mac) bordering enteric ganglia show constitutive MHC II expression in all treatment conditions (scale bar: 50µm for all images). (E) Orthogonal views of confocal z-stacks confirm MHC II (red) labeling in GFAP-expressing (grey) enteric glia (scale bar: 50µm). (F) Line-scans through enteric ganglia demonstrate co- expression of MHC II (red) and GFAP (blue) labeling. (G) Quantified mean fluorescent intensity of MHC II labeling on GFAP-expressing enteric glia in the myenteric plexus (n= 7-11 mice; 2-way analysis of variance with Tukey-corrected multiple comparisons; *P ≤ .05, ****P ≤ .0001). (H) Representative confocal images of GFAP (grey), MHC II (red) and CD74 (cyan) immunolabeling in wildtype animals treated with IFNγ and LPS show a neuronal and glial distribution of CD74 (scale bar: 50µm for all images). (I) Line-scans through enteric ganglia show minimal overlap between MHC II (red) and CD74 (green) in GFAP-expressing (blue) enteric glia. 106 Figure 3.2: Exposure to IFNγ and LPS causes weight loss but does not cause significant tissue damage or observable inflammation. (A) Animals show weight loss when treated with IFNγ + LPS in wildtype (P = .0054) and knockout (P < .0001) animals (n=5-10 mice; 2-way analysis of variance with Tukey-corrected multiple comparisons) (B) Sickness scores were not significantly different between IFNγ + LPS challenged animals compared to controls. (C) No tissue architecture changes were 107 Figure 3.2 (cont’d): observed in any experimental group. (D) No elevation in tissue histology scores was present in any condition. 108 Enteric glia express mature MHC II in response to IFNγ + LPS Enteric glia increased MHC II expression in animals challenged with IFNγ and LPS despite no overt inflammation or tissue architecture changes (Fig.3.1d; Fig.3.2). Within enteric ganglia, MHC II expression is limited to glia and no neuronal expression was observed (Fig.3.1d). Confocal image analyses confirm that MHC II primarily colocalizes with GFAP-labeled enteric glia in enteric ganglia (Mander’s overlap coefficient of 90%; Fig.3.1e-f). Treatment with IFNγ alone was not sufficient to induce glial MHC II expression (Fig.3.1d). Quantification of MHC II immunolabeling shows that enteric glia significantly (P = .0370) upregulate MHC II expression in response to IFNγ and LPS in wildtype animals, and that this effect is absent in animals that lack glial MHC II (Sox10CreERT2;IABfl/fl) (Fig.3.1g). Muscularis macrophages at the level of the myenteric plexus demonstrate constitutive expression of MHC II (54), and exposure to IFNγ and LPS stimuli did not alter expression level or pattern in wildtype or glial MHC II KO animals (Fig.3.3a,b). These observations are consistent with prior data showing that LPS stimulates enteric glial MHC II expression through TLR4 receptor pathways (62, 70) and that enteric neurons lack expression of TLR4 and MHC II (19). Importantly, these results show that the MHC II KO mouse model effectively ablates MHC II expression specifically in enteric glia and not within immune cells, thereby isolating any changes in immune activation to enteric glia. In antigen presenting cells, immature MHC II molecules contain CD74 bound within their peptide-binding groove until fusion with phagolysosomes when the CD74 dissociates to allow loading of processed peptides (58, 60). We tested whether the MHC II molecules expressed by enteric glia are mature by labeling for CD74. CD74 109 immunoreactivity was present in both enteric neurons and glia (Fig.3.1h) and this is consistent with the pattern of CD74 gene expression in enteric neurons and glia (ADD REFS for databases here). However, no co-labeling of enteric glial MHC II and CD74 was detected (Fig.3.1h). Furthermore, line-scans through enteric ganglia show little colocalization between CD74 and MHC II (Mander’s overlap coefficient of 2%; Fig.3.1i). Together, these results show that enteric glia are sensitive to pro-inflammatory stimuli and respond to low doses of IFNγ and LPS by expressing mature MHC II molecules. 110 Figure 3.3: Pattern and level of MHC II expression in macrophages does not change when treated with IFNγ and LPS. (A) Representative confocal images of MHC II (red) immunofluorescent labeling in enteric ganglion (scale bar: 50µm for all images). (B) Quantified mean fluorescent intensity of MHC II labeling in muscularis macrophages bordering enteric ganglion (n = 4-5 mice; 2-way analysis of variance with Tukey-corrected multiple comparisons). 111 Enteric glial MHC II expression contributes to CD4+ T-cell activation in mesenteric lymph nodes MHC II molecules expressed by professional antigen presenting cells play an important role in interactions that activate T-lymphocytes. Therefore, we tested whether glial MHC II molecules are functionally relevant in antigen presentation to T- lymphocytes. We began by using immunolabeling to examine whether enteric glia and T-lymphocytes are within sufficient proximity for potential interactions. In support, CD4 + T-lymphocytes were observed in close proximity to enteric glia at the level of the myenteric plexus (Fig.3.4). Next, we tested whether glial MHC II mediates functional interactions with T-lymphocytes that influence their activation state by measuring CD69 expression in T-lymphocytes in control and glial MHC II KO animals. The majority of CD4+ cells are in an inactive state in wildtype control animals and only 38% of CD4+ cells expressed the acute activation marker CD69 (Fig.3.5b,c). Exposure to IFNγ and LPS expanded the population of activated CD4+ cells by 29% and increased the percentage of CD4+ cells expressing CD69 to 67% (P = .0010). Ablating glial MHC II significantly (P = .0309) blunted the effect of IFNγ and LPS and decreased the proportion of CD4+ cells expressing CD69 to 46%, which is similar to the proportion of active CD4+ cells observed in saline treated control animals (Fig.3.5c). Glial MHC II ablation had no effect on the percentage of activated CD4+ cells in saline treated control animals (38%)(Fig.3.5b,c), and this is expected given that glial MHC II expression is only observed in animals challenged with LPS (Fig.3.1) . Enteric glial MHC II expression also affected CD8+ T-lymphocyte activation in mesenteric lymph nodes (Fig.3.6a). In healthy wildtype control animals, 61% of CD8+ T-lymphocytes 112 expressed CD69 and this proportion significantly increased to 87% in wildtype animals challenged with IFNγ and LPS (P = .0132) (Fig.3.6b). However, the IFNγ and LPS challenge did not affect CD8+ T-lymphocyte activation in KO animals (Fig.3.6b). Together, these results show that enteric glial antigen presentation mediated by MHC II plays an important role in regulating immune activity through effects on CD4+ and CD8+ T-lymphocytes. Since enteric glial MHC II ablation influences the activation state of CD4+ T- lymphocytes, we examined whether a specific T-lymphocyte subtype is preferentially affected by glial MHC II. The baseline activated proportions of T-lymphocyte subtypes expressing CD69 in wildtype control animals were 66% of IFNγ+ cells, 48% of IL4+ cells, 46% of IL17+ cells, and 43% of FoxP3+ cells (Fig.3.5d, Fig.3.7). Challenge with IFNγ and LPS in wildtype animals did not change the activated portion of IFNγ+ or IL4+ cells, but significantly elevated the activated portions of IL17+ (P = .0025) and FoxP3+ (P = .0121) cells to 83% and 77%, respectively (Fig.3.5d, Fig.3.7). Glial MHC II ablation in stimulated animals did not significantly alter the activated portions of IFNγ+ or IL4+ cells, but significantly decreased the activated portions of IL17+ (P = .0021) and FoxP3+ (P = .0057) cells to levels comparable to healthy wildtype controls. Glial MHC II ablation in control animals had no effect on T-lymphocyte subtype activation. Taken together, these results show that enteric glial MHC II primarily affects the activation state of Th17 and Treg subtypes within the CD4+ population of T-lymphocytes in mesenteric lymph nodes. Given the major effect of glial MHC II ablation on T-lymphocyte activation in mesenteric lymph nodes, we wondered if local Sox10+ cells might exist within 113 mesenteric lymph nodes that would be susceptible to MHC II ablation and mediate the observed effects. We assessed this possibility by using Sox10CreERT2+/-;tdTomato reporter mice to screen for potential Sox10+ cells in the mesenteric lymph nodes. We did not detect any tdTomato-expressing Sox10+ cells within mesenteric lymph nodes in either resting conditions or when stimulated with IFNγ and LPS (data not shown). The absence of Sox10+ cells in mesenteric lymph nodes suggests that changes in T- lymphocyte activation state are due to changes in enteric glial MHC II expression in the intestine and not other cell types residing in the mesenteric lymph node. 114 Figure 3.4: CD4 expressing T-lymphocytes are in close proximity with GFAP expressing enteric glia in the myenteric plexus. Representative image of GFAP (grey) and CD4 (red) immunofluorescent labeling in a colon cross section. Enlarged image emphasizes the presence of CD4+ T-lymphocytes within the myenteric plexus. The top region of image represents mucosal surface and bottom represents serosal surface (scale bar: 50µm) 115 Figure 3.5: Enteric glial MHC II expression affects T-lymphocyte activation. (A) Schematic depicting enteric glial cells expressing MHC II when stimulated with IFNγ and LPS. MHC II molecules interact with T-lymphocytes and cause changes in activation state as measured by CD69 expression. (B) Representative flow cytometric contour plots of mesenteric lymph nodes labeled with CD4 and CD69. Proportion of total gated cells in each quadrant expressed as percentage. (C) Levels of activated CD4+ cells quantified as a percentage of total CD4+ cells that are CD4+CD69+ (n=8-10 mice; 2- 116 Figure 3.5 (cont’d): way analysis of variance with Tukey-corrected multiple comparisons; *P ≤ .05, ***P ≤ .001). (D) Levels of activated IFNγ+, IL-4+, IL-17+, or FoxP3+ cells quantified as a percentage of total subtype-marker expressing cells that are also expressing CD69 (n=4-7 mice; 2-way analysis of variance with Tukey-corrected multiple comparisons; *P ≤ .05, **P ≤ .005). 117 Figure 3.6: Activation of CD8+ T-lymphocytes decreased when MHC II ablated in enteric glia. (A) Representative contour plots of mesenteric lymph nodes labeled with CD8 and CD69. Proportion of gated cells in each quadrant expressed as percentage. (B) Proportion of total CD8+ T-lymphocytes expressing CD69 quantified in each treatment condition (n=3-4 mice; 2-way analysis of variance with Tukey-corrected multiple comparisons; * P ≤ .05). 118 Figure 3.7: Enteric glial MHC II expression affect FoxP3+ and IL-17+ cell populations to a greater degree than IFNγ+ and IL-4+ cell populations. Representative contour plots of mesenteric lymph nodes labeled with FoxP3, IFNγ, IL-4, IL-17, and CD69. Proportion of total gated cells in each quadrant expressed as percentage. 119 Enteric glia affect local, but not serum or colonic cytokine levels Enteric glia express pro-inflammatory cytokines when stimulated with IFNγ and LPS in vitro (45). We tested whether IFNγ and LPS stimulation in wildtype animals in vivo causes enteric glial gene expression of select cytokines. Gene expression in majority of cytokines examined were not significantly affected by IFNγ and LPS challenge. However, a significant (P = .0001) increase in IL-1 gene expression and a trend towards increased tumor necrosis factor (P = .0595) was observed (Fig.3.8). Our data suggests that enteric glia respond to in vivo IFNγ and LPS challenge by altering gene expression to select cytokines and may alter cytokine levels in the local microenvironment. Activation of T-lymphocytes during inflammation has profound effects on tissue and serum cytokine levels (64, 82). Since enteric glial MHC II expression alters T- lymphocyte activation, we examined whether enteric glial MHC II expression alters cytokine levels as a downstream effect. The majority of molecules examined in a panel of 31 cytokines and chemokines were not significantly affected by the IFNγ and LPS stimulus (Fig.3.9b,c). However, significant increases were observed in colonic tissue levels of leukemia inhibitory factor and CCL5 (Fig.3.10), and serum levels of granulocyte colony stimulating factor, macrophage colony stimulating factor, IL-6, CXCL-1, and CCL4 (Fig.3.11). Ablating enteric glial MHC II had little effect on colonic tissue and serum cytokine levels and expression in MHC II KO animals did not differ significantly from wildtype animals (Fig.3.10, 3.11). Therefore, while glial antigen presentation does affect the activation profiles of immune cells in mesenteric lymph nodes, serum and regional cytokine profiles are mainly driven by other immune cells in this model. 120 It is possible that the lack of effects on serum cytokines in Sox10CreERT2;IABfl/fl mice is complicated by also ablating MHC II in Sox10+ glia in the spleen (73). In agreement with prior work, we observed that Sox10+ spleen glia do express MHC II (Fig.3.12). However, Sox10 expressing cells in the spleen were rare and their calculated density is only 1.5 cells / mm2 in splenic tissue sections (data not shown). Therefore, it is unlikely that MHC II ablation in Sox10+ cells of the spleen has a major impact that would confound our cytokine measurements in the serum. 121 Figure 3.8: Enteric glial gene expression. Gene expression of select cytokines / 122 Figure 3.8 (cont’d): chemokines in RiboTag animals stimulated with IFNγ + LPS or controls (n = 2-6 mice; unpaired student’s t-test; **P ≤ .005, ***P ≤ .0005). 123 Figure 3.9: Enteric glial MHC II expression has limited effects on serum and colonic tissue cytokine levels. (A) Schematic depicting potential source of cytokines release from T-lymphocytes after activation via binding enteric glial MHC II. (B) Heat map depicting fold change compared to saline controls in quantified cytokines and chemokines in 124 Figure 3.9 (cont’d): colonic tissue homogenates of male mice (n=4-10 mice). (C) Heat map depicting fold change compared to saline controls in quantified cytokines and chemokines in serum samples of male mice (n=4-8 mice). 125 Figure 3.10: MHC II ablation in enteric glial cells do not show changes in colonic tissue level of select cytokines. Enteric glial MHC II KO animals challenged with IFNγ 126 Figure 3.10 (cont’d): + LPS do not display significantly lower levels of regional colonic cytokines compared to wildtype counterparts challenged with IFNγ + LPS. 127 Figure 3.11: MHC II ablation in enteric glial cells do not show changes in serum level of select cytokines. Enteric glial MHC II KO animals challenged with IFNγ + LPS 128 Figure 3.11 (cont’d): do not display significantly lower levels of serum cytokines compared to wildtype counterparts challenged with IFNγ + LPS. 129 Figure 3.12: Sox10 expressing cells are present in sparse numbers in spleen tissue and express MHC II. (A) Representative confocal images of tdTomato (white) 130 Figure 3.12 (cont’d): and MHC II (red) immunofluorescent labeling in spleen sections in IFNγ + LPS treated and control animals (scale bar: 50µm for all images). (B) Quantified MHC II mean fluorescent intensity in Sox10tdTomato expressing cells (C-D) Line-scan of MHC II (red) and Sox10tdTomato (blue) labeled spleen tissue from (C) control or (D) IFNγ + LPS treated mice. 131 Pro-inflammatory stimuli IFNγ and LPS do not trigger phagocytosis in enteric glia The effects of glial MHC II on T-lymphocyte activation state suggest that glia MHC II molecules are not empty and are loaded with peptides(63, 75). In professional antigen presenting cells, peptides loaded onto MHC II are typically derived from the phagocytosis and processing of foreign antigens (60). We tested whether enteric glia present phagocytosed antigens onto MHC II molecules by first examining the phagocytic potential of enteric glia. Exposure to pHrodo BioParticles elicited phagocytosis in CD45+ immune cells neighboring enteric ganglia (Fig.3.13c), but phagocytosis or internalization of labeled E.coli particles was never observed in tdTomato+ enteric glia in 18 separate experiments (Fig.3.13b,c). We reasoned that this lack of effect might be due to using pHrodo labeled E.coli particles as the phagocytic substrate and that glia might engulf material through other mechanisms such as endocytosis. Therefore, we used labeled LPS molecules and 1.0µm fluorescent polystyrene microspheres to test whether TLR4-mediated endocytosis or size are contributing factors to glial phagocytosis. Incubation with fluorescently labeled LPS or fluorescent polystyrene microspheres did not elicit any uptake within enteric glial cells (data not shown), suggesting that enteric glia are either incapable of phagocytosis or that there is insufficient stimulation to trigger phagocytic behavior in enteric glia. Taken together, our results demonstrate that expression of MHC II molecules on enteric glia when stimulated with low dose IFNγ and LPS is not due to the phagocytosis of foreign antigens but is derived from other pathways. 132 Figure 3.13: Enteric glia do not exhibit phagocytic activity when stimulated with IFNγ and LPS. (A) Schematic depicting that enteric glial cells do not exhibit phagocytic activity when stimulated with IFNγ and LPS. (B) Representative brightfield and epifluorescent image of tdTomato-expressing Sox10+ cells (blue) and pHrodo labeled E.coli BioParticles (green). (C) Representative confocal image of tdTomato-expressing Sox10+ cells (blue), pHrodo labeled E.coli BioParticles (green), and CD45 immunolabeling (magenta) (scale bar: 50µm). 133 Glial autophagy regulates MHC II expression Our data show that glial MHC II expression occurs independently of phagocytosis. Therefore, we examined alternative pathways for MHC II induction. In antigen presenting cells such as B cells, a deficiency in autophagic protein Atg5 decreases MHC II expression levels, suggesting that autophagy regulates MHC II expression levels in antigen presenting cells (78). We tested whether a similar autophagy mechanism controls enteric glial MHC II expression by measuring levels of the autophagy proteins p62 and LC3 over a 16-hour time-course. As described above, MHC II expression was detected in enteric glia by 4 hours post injection (Fig.3.14b). Expression of autophagic proteins LC3 and p62 was not limited to enteric glia and is distributed in both enteric neurons and glia. The punctate nature of LC3 labeling shows a greater density in enteric glia, while p62 labeling is more prominent in neurons compared to glia, consistent with findings in single-cell RNA seq databases (19) (Fig.3.14b,c). Glial MHC II expression significantly (P = .0021) increased by 8 hours post-injection with IFNγ and LPS and further increased by 16 hours (P < .0001) (Fig. 3.14b,c,d). LC3+ puncta density in both enteric glia (Fig.3.14f) and enteric neurons (Fig.3.14i) trended towards increased levels over 16 hours, while p62 expression was significantly elevated in enteric glia (P = 0.0060) and neurons (P = .0270) by 4 hours post-injection and subsequently decreased to pre-injection levels by 16 hours (Fig.3.14e,h). The spike and subsequent decrease in p62 labeling, with the concurrent trend in increasing LC3 puncta over 16 hours suggests that autophagy is active in both enteric glia and neurons (55). The timing of MHC II upregulation in enteric glia coincides with autophagy-mediated consumption of p62 molecules and accumulation of LC3 134 deposits, showing that enteric glial MHC II expression occurs concurrently with autophagy pathways. Since autophagy can lead to the presentation of intracellular antigens on MHC II molecules (18), we tested whether our findings are indicative of autophagy-induced glial MHC II expression. We used a combination of IFNγ and LPS, the autophagy stimulator rapamycin, and the autophagy inhibitor 3-methyladenine (3-MA) to test the regulation of glial MHC II expression by autophagy. A 16-hour incubation with IFNγ and LPS significantly (P = 0.0015) increased MHC II levels in enteric glia, similar to our in vivo findings (Fig.3.14g). Combining IFNγ and LPS with rapamycin further increased glial MHC II expression level compared to control (P < .0001), but interestingly, incubation with rapamycin alone did not increase MHC II levels compared to control (P = .8354) (Fig.3.14g). However, tissues incubated with a combination of IFNγ, LPS, and 3-MA, or IFNγ, LPS, rapamycin, and 3-MA had significantly decreased levels of glial MHC II expression compared to non-3-MA treated counterparts (P = .0047 and P < .0001, respectively) and reduced glial MHC II expression to levels comparable to untreated controls (Fig.3.14g). Taken together, our data show that inhibition of glial autophagy effectively eliminates MHC II labeling, suggesting that glial MHC II expression requires autophagy activation. However, the inability for rapamycin alone to induce glial MHC II suggests that the activation of autophagy alone is insufficient to trigger glial MHC II expression. 135 Figure 3.14: Enteric glia express MHC II via autophagy pathways. (A) Schematic depicting enteric glial MHC II loading 136 Figure 3.14 (cont’d): and expression of autophagy-derived peptides when stimulated with IFNγ and LPS. (B) Representative confocal images of GFAP (grey), MHC II (red), and LC3 (cyan) immunofluorescent labeling in enteric ganglia in wildtype animals injected with IFNγ and LPS over a 16-hour time-course. LC3 show punctate labeling and is distributed in a neuronal and glial pattern (scale bar: 50µm for all images). (C) Representative confocal images of GFAP (grey), MHC II (red), and P62 (cyan) immunofluorescent labeling in enteric ganglia in wildtype animals injected with IFNγ and LPS over a 16-hour time- course. P62 labeling is prominent in enteric neurons but is distributed in both neurons and glia (scale bar: 50µm for all images). (D,E) Quantified mean fluorescent intensity of MHC II and p62 labeling on GFAP-expressing enteric glia in the myenteric plexus. Wildtype animals were injected with IFNγ and LPS and colons harvested over a 16-hour time-course ((D) n=5-8 mice and (E) n=3-4 mice; 1-way analysis of variance with Sidak- corrected multiple comparisons; *P ≤ .05, **P ≤ .005, ****P ≤ .0001). (F) Quantified density of LC3 puncta labeling over the area of GFAP-expressing enteric glia in the myenteric plexus. Wildtype animals were injected with IFNγ and LPS and colons harvested over a 16-hour time-course (n=2-3 mice; 1-way analysis of variance with Sidak-corrected multiple comparisons). (G) Quantified mean fluorescent intensity of MHC II labeling on GFAP-expressing enteric glia in the myenteric plexus. LMMP tissue preparations from wildtype animals were incubated with a combination of IFNγ and LPS, Rapamycin, and 3-MA for 16 hours prior to fixation and immunolabeling. (n=4-5 mice; 1- way analysis of variance with Sidak-corrected multiple comparisons; **P ≤ .005, ****P ≤ .0001). (H) Quantified mean fluorescent intensity of p62 labeling within enteric ganglion 137 Figure 3.14 (cont’d): outside of GFAP-expressing enteric glia in the myenteric plexus. Wildtype animals were injected with IFNγ and LPS and colons harvested over a 16-hour time-course (n=3-4 mice; 1-way analysis of variance with Sidak-corrected multiple comparisons). (I) Quantified density of LC3 puncta labeling within enteric ganglion outside of GFAP- expressing enteric glia in the myenteric plexus. Wildtype animals were injected with IFNγ and LPS and colons harvested over a 16-hour time-course (n=2-3 mice; 1-way analysis of variance with Sidak-corrected multiple comparisons). 138 Discussion The notion that enteric glia function as antigen presenting cells stems from observations in Crohn’s and Chagas disease where glia increase MHC II expression (REFS). The functional significance of these observations has remained in question ever since. Here, we show that antigen presentation by enteric glia functions to modulate immune responses through interactions with T-lymphocytes. Enteric glia are sensitive to inflammatory stimuli and respond to low dose IFNγ and LPS by expressing MHC II molecules. Enteric glial MHC II molecules are functional and contribute to CD4+ T-lymphocyte activation, particularly in the Th17 and Treg subpopulations. These effects do not involve large shifts in proinflammatory mediators. Glial MHC II expression is not phagocytosis-mediated and is instead, dependent on autophagy activation. However, autophagy activation alone is not sufficient to elicit glial MHC II expression. MHC II is primarily expressed by professional antigen presenting cells and plays an important role in their interactions with T-lymphocytes. We show that enteric glia express MHC II in an inducible manner when exposed to proinflammatory stimuli, consistent with prior in vitro and in vivo human studies which show that glial MHC II expression is induced by enteroinvasive E.coli or inflammation (3, 23, 70). Furthermore, the punctate distribution of MHC II along glial processes is consistent with findings in Chagas disease (3). In our study, enteric glia expressed MHC II in the absence of inflammation or observable tissue damage. This suggests that glial antigen presentation may act as an early responder to inflammatory stress. This conclusion is consistent with observations in Crohn’s disease showing that enteric glia express MHC II in diseased areas and macroscopically uninvolved areas (23). Interestingly, occasional glial MHC II 139 labeling is also observed in samples from healthy individuals (3, 23). We did not observe glial MHC II expression in healthy animals and it is not observed in cultured human enteric glia in vitro (70). This discrepancy in baseline glial MHC II expression may be due to the lack of a microbiome in vitro, and potentially due to murine tolerance to LPS and bacterial components (14). However, baseline glial expression of MHC II in humans suggests that enteric glia constantly monitor the ENS and modulate immune activation in response to the constant presence of microbiome-derived antigens. The baseline MHC II expression also suggests that enteric glial MHC II may have anti- inflammatory effects. Anti-inflammatory actions of MHC II may contribute to homeostasis by preventing repeated immune activation and inflammation against the normal microbiome. Similar anti-inflammatory effects of MHC II expression by intestinal epithelial cells limit Th1 cell infiltration and modulate T-lymphocyte subtype distribution in the intestine (69). The presence of enteric glial MHC II expression in vivo and in vitro (3, 23, 70) suggests potential interactions with T-lymphocytes. Enteric glial interactions with T- lymphocytes in vivo when expressing neoantigens (15) suggest that T-lymphocytes can recognize enteric glia. Moreover, enteric glia inhibit T-lymphocyte activation in co- cultures (36). However, the functional role of antigen presentation by enteric glia and their potential to influence T-lymphocytes in vivo is not understood. Here, we show that enteric glial MHC II expression plays an important role in the activation of CD4+ T- lymphocytes in mesenteric lymph nodes. We also noted a surprisingly large population of CD4- cells expressing CD69 in stimulated animals. A portion of those cells responding to glial MHC II represent CD69-expressing CD8+ T-lymphocytes (51, 57, 140 59). The decreased CD8+ T-lymphocyte activation may contribute to the lack of histological tissue damage. Additional cell types may include natural killer cells (11, 33) and dendritic cells (2, 40), but further study is warranted to further elucidate this cell population. Our data show that glial MHC II expression has a propensity towards affecting Th17 and Treg subpopulations moreso than Th1 and Th2 subpopulations. This apparent conflict in immune regulation by activating both pro- and anti-inflammatory T- lymphocyte subpopulations suggest a feedback inhibition to limit Th17 activity (9). Alternatively, the Th17 subpopulation may exhibit anti-inflammatory actions by secreting anti-inflammatory cytokines (22, 52, 56, 84). Furthermore, enteric glia may contribute to an anti-inflammatory phenotype by destabilizing interactions with certain T-lymphocyte subtypes to inhibit proliferation, similar to effects seen in dendritic cells (76). Despite enteric glial MHC II expression having no apparent effect on regional tissue cytokine levels, gene expression changes suggest that enteric glia may have a mixed role in the local ENS microenvironment by expressing IL-1 and tumor necrosis factor, while decreasing IL-12b. The apparent expression of certain pro-inflammatory cytokines despite decreasing others may be due to the concurrent activation of separate pathways. While IL-1 and tumor necrosis factor gene expression may be driven by direct binding of LPS molecules onto TLR4 receptors on enteric glia, a decrease in IL- 12b may be due to the activation of other separate signaling pathways. In support, enteric glia show strong immunosuppressive effects that inhibit T-lymphocyte proliferation in co-culture models and this effect is strengthened in enteric glia derived from Crohn’s patients (36). In contrast, enteric glia can contribute to neuronal cell death (7), and cultured human enteric glia challenged with IFNγ and LPS increase gene 141 expression of various pro-inflammatory cytokines (45). This suggests that the dynamic immunomodulatory capabilities of enteric glia depend on the surrounding environment. Our current data focuses on the glial MHC II KO (Sox10CreERT2;IABfl/fl) model and its effects on T-lymphocytes of the mesenteric lymph nodes. Since we did not detect any Sox10+ cells that would be susceptible to the MHC II KO in mesenteric lymph nodes, how enteric glia in the ENS affect T-lymphocyte activation while in separate anatomical locations remains to be understood. Given the proximity of local CD4+ T- lymphocytes to enteric glia, it is possible that enteric glia influence the activation of local T-lymphocytes via cell interactions or secreted compounds, and the migration of activated lymphocytes could subsequently have a wider reaching effect in regional lymph nodes (36). Similarly, enteric glia may potentially influence nearby dendritic cells to bring about lymph node effects. Enteric glial expression of 15-deoxy-Δ12,14- prostaglandin J2 (1) can conceivably influence dendritic cells to stimulate activation of type 1 regulatory T-lymphocytes (72), but this interaction has yet to be described. Phagocytosis is an important pathway for professional antigen presenting cells to uptake foreign antigens for processing and presentation. In our study, we did not find evidence that enteric glia uptake labeled E.coli, labeled LPS, or fluorescent latex beads. The lack of phagocytosis in our study, however, may be due to the strength of the proinflammatory stimuli used, since others have successfully demonstrated phagocytic potential in glia in other organ systems (49) and in other related glial populations in the CNS (21, 34, 38, 71). Therefore, stronger stimuli or the dysfunction and death of nearby cells may be necessary for enteric glia to demonstrate phagocytic activity. Given their shared characteristics with astrocytes (12), their proximity to enteric neurons (5) and 142 their ability to contribute to neuronal demise (7), it is conceivable that enteric glia also uptake neuronal fragments during neuronal injury or death. The presentation of neuronal fragments to the adaptive immune system could lead to the generation of autoreactive antibodies and may be a mechanism contributing to the production of anti- HuC/D antibodies in patients with irritable bowel syndrome (77). The expression of enteric glial MHC II in situations of inflammation and neuron loss, such as in Chagasic megacolon or Crohn’s disease (3, 23), suggest that enteric glia play a role in these disease states. Whether enteric glia contribute to neuronal death and inflammation through MHC II expression or promote anti-inflammatory pathways to maintain homeostasis remains to be studied. The ability of enteric glia to express MHC II molecules in the absence of phagocytosis suggests that glia, like B-lymphoma cell lines, use alternative pathways such as autophagy promote the loading of intracellular and self-antigens onto MHC II for presentation (18, 67). Enteric glial MHC II expression is dependent on autophagy activation and the timing of MHC II expression coincides with autophagy activation, which raises the possibility that enteric glia present autophagy-derived peptides on their MHC II molecules. Although the presentation of autophagy-derived self-antigens on MHC II molecules suggest the potential development of autoimmune reactions, our findings suggest otherwise, as no pro-inflammatory cytokine levels were changed when glial MHC II expression status changed and no inflammatory cell recruitment was observed in colonic tissue. These data are consistent with findings by other groups demonstrating anti-inflammatory effects from autophagy and autophagy-induced MHC II 143 expression (76, 81). However, mass spectrometry analysis of glial MHC II molecules is necessary to conclusively determine the identity of presented peptides. The autophagic response to IFNγ and LPS differed between enteric neurons and glia in their expression of LC3 and p62, with more prominent labeling of p62 in enteric neurons and greater LC3 puncta in enteric glia. Although unclear, the more prominent p62 labeling may indicate that enteric neurons exhibit a higher sensitivity to inflammatory stimuli than enteric glia. The observed elevation in density of glial LC3 puncta may in fact be related to glial phagocytosis and antigen presentation in the form of LC3-associated phagocytosis via TLR4 activation (25, 26, 61). However, as we did not detect any phagocytic activity, a more detailed examination specifically into LC3- associated phagocytosis is needed to test this hypothesis (50). Taken in the context of IBD, which is considered having dysfunctional autophagy pathways (32, 41), the activation of autophagy in enteric glia and neurons may in fact be a compensatory mechanism to limit potential inflammatory damage to the ENS. This potential mechanism is indeed supported by findings from numerous groups demonstrating the activation of autophagy pathways can inhibit intestinal inflammation and reduce colitis (37, 47, 81). Our data show that autophagy activation is required for enteric glial MHC II expression, agreeing with prior studies demonstrating that dendritic cells require autophagy machinery to express peptides on MHC II molecules (43). Furthermore, our data show that autophagy activation by rapamycin alone is insufficient to induce glial MHC II expression. This may be due to the nature of rapamycin, as other groups have shown in vitro that rapamycin has varying effectiveness in eliciting autophagy in 144 different glial cell lines (68). Alternatively, enteric glia may simply require both a stimulus with a concomitant activation in autophagy to express MHC II molecules. However, LPS as a stimulus may in fact contribute to autophagy inhibition (44, 83). Taken together with the upregulation of pro-inflammatory cytokine gene expression, our data suggest that two separate pathways may be concurrently activated by IFNγ and LPS challenge. Direct LPS binding onto enteric glial TLR4 receptors may contribute to a pro- inflammatory phenotype in enteric glia, while the activation of a separate pathway induces autophagy and an anti-inflammatory phenotype. In conclusion, our results show that enteric glia respond to inflammatory stimuli by expressing functional MHC II molecules to modulate T-lymphocyte activation (Fig.3.15). Furthermore, we show that autophagy activation is necessary, but insufficient, to induce enteric glial MHC II expression. Together, these observations provide evidence for enteric glia being an important immune regulator during inflammatory stress and suggest that they may play an anti-inflammatory role during intestinal inflammation and IBD in an attempt to maintain homeostasis. 145 Figure 3.15: Schematic mechanism of enteric glial MHC II expression. Enteric glia exposed to pro-inflammatory stimuli IFNγ and LPS upregulate expression of MHC II molecules on their cell surface. Enteric glial MHC II is not derived from phagocytosed foreign antigens, but rather derived from products of autophagy. Enteric glial MHC II are functional molecules that can interact with CD4+ T-lymphocytes and modulate CD69 expression levels primarily in IL-17 and FoxP3 expressing CD4+ T-lymphocytes. 146 REFERENCES 147 REFERENCES 1. Abdo H, Mahé MM, Derkinderen P, Bach-Ngohou K, Neunlist M, Lardeux B. The omega-6 fatty acid derivative 15-deoxy-Δ12,14-prostaglandin J2 is involved in neuroprotection by enteric glial cells against oxidative stress. J Physiol 590: 2739–50, 2012. 2. Alari-Pahissa E, Notario L, Lorente E, Vega-Ramos J, Justel A, López D, Villadangos JA, Lauzurica P. CD69 Does Not Affect the Extent of T Cell Priming. PLoS One 7, 2012. 3. Barcelos Morais Da Silveira A, De Oliveira EC, Neto SG, Luquetti AO, Toshio Fujiwara R, Correa Oliveira R, Brehmer A, Barcelos A, Oliveira EC De, Neto SG, Luquetti AO, Toshio R, Correa R, Brehmer A. Enteroglial cells act as antigen-presenting cells in chagasic megacolon. Hum Pathol 42: 522–532, 2011. 4. Birnboim HC, Lemay A-M, Lam DKY, Goldstein R, Webb JR. Cutting edge: MHC class II-restricted peptides containing the inflammation-associated marker 3- nitrotyrosine evade central tolerance and elicit a robust cell-mediated immune response. J Immunol 171: 528–32, 2003. 5. Boesmans W, Martens MA, Weltens N, Hao MM, Tack J, Cirillo C, Vanden Berghe P. Imaging neuron-glia interactions in the enteric nervous system. Front Cell Neurosci 7: 183, 2013. 6. Bolte S, Cordelières FP. A guided tour into subcellular colocalization analysis in light microscopy. J. Microsc. 224 Blackwell Publishing Ltd: 213–232, 2006. 7. Brown IAM, McClain JL, Watson RE, Patel BA, Gulbransen BD. Enteric Glia Mediate Neuron Death in Colitis Through Purinergic Pathways That Require Connexin-43 and Nitric Oxide. C Cell Mol Gastroenterol Hepatol 2: 77–91, 2016. 8. Chastain EML, Duncan DS, Rodgers JM, Miller SD. The role of antigen presenting cells in multiple sclerosis. Biochim. Biophys. Acta - Mol. Basis Dis. 1812 NIH Public Access: 265–274, 2011. 9. Chaudhry A, Samstein RM, Treuting P, Liang Y, Pils MC, Heinrich J, Jack RS, Wunderlich FT, Brüning JC, Müller W, Rudensky AY. Interleukin-10 signaling in regulatory T cells is required for suppression of Th17 cell-mediated inflammation. Immunity 34: 566–78, 2011. 10. Cheadle GA, Costantini TW, Lopez N, Bansal V, Eliceiri BP, Coimbra R. Enteric Glia Cells Attenuate Cytomix-Induced Intestinal Epithelial Barrier Breakdown. PLoS One 8: 1–11, 2013. 11. Cibrián D, Sánchez-Madrid F. CD69: from activation marker to metabolic gatekeeper. Eur. J. Immunol. 47 Wiley-VCH Verlag: 946–953, 2017. 148 12. Coelho-Aguiar J de M, Bon-Frauches AC, Gomes ALT, Verissimo CP, Aguiar DP, Matias D, Thomasi BB de M, Gomes AS, Brito GA de C, Moura-Neto V. The enteric glia: Identity and functions. Glia 63: 921–935, 2015. 13. Colombo E, Farina C. Astrocytes: Key Regulators of Neuroinflammation. Trends Immunol 37: 608–620, 2016. 14. Copeland S, Warren HS, Lowry SF, Calvano SE, Remick D. Acute inflammatory response to endotoxin in mice and humans. Clin Diagn Lab Immunol 12: 60–67, 2005. 15. Cornet A, Savidge TC, Cabarrocas J, Deng W-L, Colombel J-F, Lassmann H, Desreumaux P, Liblau RS. Enterocolitis induced by autoimmune targeting of enteric glial cells: A possible mechanism in Crohn’s disease? Proc Natl Acad Sci 98: 13306–13311, 2001. 16. Delvalle NM, Dharshika C, Morales-Soto W, Fried DE, Gaudette L, Gulbransen BD. Communication Between Enteric Neurons, Glia, and Nociceptors Underlies the Effects of Tachykinins on Neuroinflammation. CMGH 6: 321–344, 2018. 17. Delvalle NM, Fried DE, Rivera-Lopez G, Gaudette L, Gulbransen BD. Cholinergic activation of enteric glia is a physiological mechanism that contributes to the regulation of gastrointestinal motility. Am J Physiol - Gastrointest Liver Physiol 315: G473–G483, 2018. 18. Dengjel J, Schoor O, Fischer R, Reich M, Kraus M, Müller M, Kreymborg K, Altenberend F, Brandenburg J, Kalbacher H, Brock R, Driessen C, Rammensee H, Stevanovic S. Autophagy promotes MHC class II presentation of peptides from intracellular source proteins. Proc Natl Acad Sci U S A 102: 7922– 7, 2005. 19. Drokhlyansky E, Smillie CS, Van Wittenberghe N, Ericsson M, Griffin GK, Eraslan G, Dionne D, Cuoco MS, Goder-Reiser MN, Sharova T, Kuksenko O, Aguirre AJ, Boland GM, Graham D, Rozenblatt-Rosen O, Xavier RJ, Regev A. The Human and Mouse Enteric Nervous System at Single-Cell Resolution. Cell 182: 1606-1622.e23, 2020. 20. Fagbemi AO, Torrente F, Puleston J, Lakhoo K, James S, Murch SH. Enteric Neural Disruption in Necrotizing Enterocolitis Occurs in Association With Myenteric Glial Cell CCL20 Expression. J Pediatr Gastroenterol Nutr 57: 788– 793, 2013. 21. Fricker M, Oliva-Martín MJ, Brown GC. Primary phagocytosis of viable neurons by microglia activated with LPS or Aβ is dependent on calreticulin/LRP phagocytic signalling. J Neuroinflammation 9: 1–12, 2012. 22. Gagliani N, Amezcua Vesely MC, Iseppon A, Brockmann L, Xu H, Palm NW, 149 de Zoete MR, Licona-Limón P, Paiva RS, Ching T, Weaver C, Zi X, Pan X, Fan R, Garmire LX, Cotton MJ, Drier Y, Bernstein B, Geginat J, Stockinger B, Esplugues E, Huber S, Flavell RA. Th17 cells transdifferentiate into regulatory T cells during resolution of inflammation. Nature 523: 221–5, 2015. 23. Geboes K, Rutgeerts P, Ectors N, Mebis J, Penninckx F, Vantrappen G, Desmet VJ. Major histocompatibility class II expression on the small intestinal nervous system in Crohn’s disease. Gastroenterology 103: 439–447, 1992. 24. Gulbransen BD, Sharkey KA. Purinergic Neuron-to-Glia Signaling in the Enteric Nervous System. Gastroenterology 136: 1349–1358, 2009. 25. Heckmann BL, Boada-Romero E, Cunha LD, Magne J, Green DR. LC3- Associated Phagocytosis and Inflammation. J. Mol. Biol. 429 Academic Press: 3561–3576, 2017. 26. Heckmann BL, Green DR. LC3-associated phagocytosis at a glance. J. Cell Sci. 132 NLM (Medline): 2019. 27. Hernandez S, Fried DE, Grubišić V, McClain JL, Gulbransen BD. Gastrointestinal neuroimmune disruption in a mouse model of Gulf War illness. FASEB J 33: 6168–6184, 2019. 28. Herzog J, Maekawa Y, Cirrito TP, Illian BS, Unanue ER. Activated antigen- presenting cells select and present chemically modified peptides recognized by unique CD4 T cells. Proc Natl Acad Sci U S A 102: 7928–7933, 2005. 29. Hood L, Steinmetz M, Malissen B. Genes of the major histocompatibility complex of the mouse. Annu Rev Immunol 1: 529–68, 1983. 30. Hyldig-Nielsen JJ, Schenning L, Hammerling U, Widmark E, Heldin E, Lind P, Servenius B, Lund T, Flavell R, Lee JS, Trowsdale J, Schreier PH, Zablitzky F, Larhammar D, Peterson PA, Rask L. The complete nucleotide sequence of the I-E alpha d immune response gene. Nucleic Acids Res 11: 5055– 71, 1983. 31. Ibiza S, García-cassani B, Ribeiro H, Carvalho T, Almeida L, Marques R, Misic AM, Bartow-McKenney C, Larson DM, Pavan WJ, Eberl G, Grice EA, Veiga-Fernandes H, Eberi G, Grice EA, Veiga-Fernandes H, Eberl G, Grice EA, Veiga-Fernandes H. Glial-cell-derived neuroregulators control type 3 innate lymphoid cells and gut defence. Nature 535: 440–443, 2016. 32. Iida T, Onodera K, Nakase H. Role of autophagy in the pathogenesis of inflammatory bowel disease. World J Gastroenterol 23: 1944–1953, 2017. 33. Jiang YZ, Couriel D, Mavroudis DA, Lewalle P, Malkovska V, Hensel NF, Dermime S, Molldrem J, Barrett AJ. Interaction of natural killer cells with MHC class II: Reversal of HLA-DR1-mediated protection of K562 transfectant from 150 natural killer cell-mediated cytolysis by brefeldin-A. Immunology 87: 481–486, 1996. 34. Jones RS, Minogue AM, Connor TJ, Lynch MA. Amyloid-β-induced astrocytic phagocytosis is mediated by CD36, CD47 and RAGE. J Neuroimmune Pharmacol 8: 301–11, 2013. 35. Kambayashi T, Laufer TM. Atypical MHC class II-expressing antigen-presenting cells: can anything replace a dendritic cell? Nat Rev Immunol 14: 719–30, 2014. 36. Kermarrec L, Durand T, Neunlist M, Naveilhan P, Neveu I. Enteric glial cells have specific immunosuppressive properties. J Neuroimmunol 295–296: 79–83, 2016. 37. Kökten T, Gibot S, Lepage P, D’Alessio S, Hablot J, Ndiaye NC, Busby- Venner H, Monot C, Garnier B, Moulin D, Jouzeau JY, Hansmannel F, Danese S, Guéant JL, Muller S, Peyrin-Birouleta L. TREM-1 inhibition restores impaired autophagy activity and reduces colitis in mice. J Crohn’s Colitis 12: 230– 244, 2018. 38. Konishi H, Okamoto T, Hara Y, Komine O, Tamada H, Maeda M, Osako F, Kobayashi M, Nishiyama A, Kataoka Y, Takai T, Udagawa N, Jung S, Ozato K, Tamura T, Tsuda M, Yamanaka K, Ogi T, Sato K, Kiyama H. Astrocytic phagocytosis is a compensatory mechanism for microglial dysfunction. EMBO J. ( September 22, 2020). doi: 10.15252/embj.2020104464. 39. Lafuse WP, Singh S, Savarirayan S, David CS. DNA restriction fragment analysis of E alpha genes in E-negative H-2 recombinant mouse strains. Immunogenetics 24: 68–70, 1986. 40. Lamana A, Martin P, De La Fuente H, Martinez-Mũoz L, Cruz-Adalia A, Ramirez-Huesca M, Escribano C, Gollmer K, Mellado M, Stein J V., Rodriguez-Fernandez JL, Sanchez-Madrid F, Del Hoyo GM. CD69 modulates sphingosine-1-phosphate-induced migration of skin dendritic cells. J Invest Dermatol 131: 1503–1512, 2011. 41. Larabi A, Barnich N, Nguyen HTT. New insights into the interplay between autophagy, gut microbiota and inflammatory responses in IBD. Autophagy 16: 38– 51, 2020. 42. Laranjeira C, Sandgren K, Kessaris N, Richardson W, Potocnik A, Vanden Berghe P, Pachnis V. Glial cells in the mouse enteric nervous system can undergo neurogenesis in response to injury. J Clin Invest 121: 3412–24, 2011. 43. Lee HK, Mattei LM, Steinberg BE, Alberts P, Lee YH, Chervonsky A, Mizushima N, Grinstein S, Iwasaki A. In vivo requirement for Atg5 in antigen presentation by dendritic cells. Immunity 32: 227–39, 2010. 151 44. Lee JW, Nam H, Kim LE, Jeon Y, Min H, Ha S, Lee Y, Kim SY, Lee SJ, Kim EK, Yu SW. TLR4 (toll-like receptor 4) activation suppresses autophagy through inhibition of FOXO3 and impairs phagocytic capacity of microglia. Autophagy 15: 753–770, 2019. 45. Liñán-Rico A, Turco F, Ochoa-Cortes F, Harzman A, Needleman BJ, Arsenescu R, Abdel-Rasoul M, Fadda P, Grants I, Whitaker E, Cuomo R, Christofi FL. Molecular Signaling and Dysfunction of the Human Reactive Enteric Glial Cell Phenotype: Implications for GI Infection, IBD, POI, Neurological, Motility, and GI Disorders. Inflamm Bowel Dis 22: 1812–1834, 2016. 46. Le Loupp A-G, Bach-Ngohou K, Bourreille A, Boudin H, Rolli-Derkinderen M, Denis MG, Neunlist M, Masson D. Activation of the prostaglandin D2 metabolic pathway in Crohn’s disease: involvement of the enteric nervous system. BMC Gastroenterol 15: 112, 2015. 47. Macias-Ceja DC, Cosín-Roger J, Ortiz-Masiá D, Salvador P, Hernández C, Esplugues J V, Calatayud S, Barrachina MD. Stimulation of autophagy prevents intestinal mucosal inflammation and ameliorates murine colitis. Br J Pharmacol 174: 2501–2511, 2017. 48. Mahdi BM. Role of HLA typing on Crohn’s disease pathogenesis. Ann Med Surg 4: 248–253, 2015. 1047–1055, 1990. 49. Mano T, Puro DG. Phagocytosis by Human Retinal Glial Cells in Culture. 31: 50. Martinez J. Detection of LC3‐Associated Phagocytosis (LAP). Curr Protoc Cell Biol 87, 2020. 51. Matechak EO, Killeen N, Hedrick SM, Fowlkes BJ. MHC class II-specific T cells can develop in the CD8 lineage when CD4 is absent. Immunity 4: 337–347, 1996. 52. McGeachy MJ, Bak-Jensen KS, Chen Y, Tato CM, Blumenschein W, McClanahan T, Cua DJ. TGF-β and IL-6 drive the production of IL-17 and IL-10 by T cells and restrain TH-17 cell-mediated pathology. Nat Immunol 8: 1390– 1397, 2007. 53. Middel P, Raddatz D, Gunawan B, Haller F, Radzun HJ. Increased number of mature dendritic cells in Crohn’s disease: Evidence for a chemokine mediated retention mechanism. Gut 55: 220–227, 2006. 54. Mikkelsen HB. Macrophages in the external muscle layers of mammalian intestines. Histol Histopathol 10: 719–736, 1995. 55. Mizushima N, Yoshimori T, Levine B. Methods in mammalian autophagy research. Cell 140: 313–26, 2010. 152 56. Musuraca G, De Matteis S, Napolitano R, Papayannidis C, Guadagnuolo V, Fabbri F, Cangini D, Ceccolini M, Giannini MB, Lucchesi A, Ronconi S, Mariotti P, Savini P, Tani M, Fattori PP, Guidoboni M, Martinelli G, Zoli W, Amadori D, Carloni S. IL-17/IL-10 double-producing T cells: New link between infections, immunosuppression and acute myeloid leukemia. J Transl Med 13: 1– 10, 2015. 57. Ranasinghe S, Lamothe PA, Soghoian DZ, Kazer SW, Cole MB, Shalek AK, Yosef N, Jones RB, Donaghey F, Nwonu C, Jani P, Clayton GM, Crawford F, White J, Montoya A, Power K, Allen TM, Streeck H, Kaufmann DE, Picker LJ, Kappler JW, Walker BD. Antiviral CD8+ T Cells Restricted by Human Leukocyte Antigen Class II Exist during Natural HIV Infection and Exhibit Clonal Expansion. Immunity 45: 917–930, 2016. 58. Riberdy JM, Avva RR, Geuze HJ, Cresswell P. Transport and intracellular distribution of MHC class II molecules and associated invariant chain in normal and antigen-processing mutant cell lines. J Cell Biol 125: 1225–1237, 1994. 59. Rist M, Smith C, Bell MJ, Burrows SR, Khanna R. Cross-recognition of HLADR4 alloantigen by virus-specific CD8+ T cells: A new paradigm for self- /nonself-recognition. Blood 114: 2244–2253, 2009. 60. Roche PA, Furuta K. The ins and outs of MHC class II-mediated antigen processing and presentation. Nat Rev Immunol 15: 203–16, 2015. 61. Romao S, Münz C. LC3-associated phagocytosis. Autophagy 10: 526–528, 2014. 62. Rosenbaum C, Schick MA, Wollborn J, Heider A, Scholz C-J, Cecil A, Niesler B, Hirrlinger J, Walles H, Metzger M. Activation of Myenteric Glia during Acute Inflammation In Vitro and In Vivo. PLoS One 11: e0151335, 2016. 63. Rupp B, Günther S, Makhmoor T, Schlundt A, Dickhaut K, Gupta S, Choudhary I, Wiesmüller KH, Jung G, Freund C, Falk K, Rötzschke O, Kühne R. Characterization of structural features controlling the receptiveness of empty class II MHC molecules. PLoS One 6, 2011. 64. Santana MA, Esquivel-Guadarrama F. Cell biology of T cell activation and differentiation. Int Rev Cytol 250: 217–74, 2006. 65. Schmid D, Pypaert M, Münz C. Antigen-loading compartments for major histocompatibility complex class II molecules continuously receive input from autophagosomes. Immunity 26: 79–92, 2007. 66. Shrum B, Anantha R V, Xu SX, Donnelly M, Haeryfar S, McCormick JK, Mele T, Bone R, Balk R, Cerra F, Dellinger R, Fein A, Knaus W, Schein R, Sibbald W, Martin G, Mannino D, Eaton S, Moss M, Vincent J, Sakr Y, Sprung C, Ranieri V, Reinhart K, Gerlach H, Moreno R, Carlet J, Gall J Le, Payen D, Levy M, Fink M, Marshall J, Abraham E, Angus D, Cook D, Cohen J, Opal S, 153 Vincent J, Ramsay G, Buras J, Holzmann B, Sitkovsky M, Marshall J, Esmon C, Nemzek J, Hugunin K, Opp M, Vincent J, Rello J, Marshall J, Silva E, Anzueto A, Martin C, Moreno R, Lipman J, Gomersall C, Sakr Y, Reinhart K, Hubbard W, Choudhry M, Schwacha M, Kerby J, Rue L, Bland K, Chaudry I, Wichterman K, Baue A, Chaudry I, Maier S, Traeger T, Entleutner M, Westerholt A, Kleist B, Huser N, Holzmann B, Stier A, Pfeffer K, Heidecke C, Dejager L, Pinheiro I, Dejonckheere E, Libert C, Knaus W, Draper E, Wagner D, Zimmerman J, Huet O, Ramsey D, Miljavec S, Jenney A, Aubron C, Aprico A, Stefanovic N, Balkau B, Head G, Haan J de, Chin-Dusting J, Langford D, Bailey A, Chanda M, Clarke S, Drummond T, Echols S, Glick S, Ingrao J, Klassen-Ross T, Lacroix-Fralish M, Matsumiya L, Sorge R, Sotocinal S, Tabaka J, Wong D, Maagdenberg A van den, Ferrari M, Craig K, Mogil J, Hyde S, Stith R, McCallum R, Gavrieli Y, Sherman Y, Ben-Sasson S, Hanley J, McNeil B, Youden W, Marshall J, Reinhart K, Lee S, Feingold D, Carter J, Zhai C, Stapleton G, Gleason N, Whelan R, Otto G, Busch M, Sossdorf M, Claus R, Hotchkiss R, Karl I, Hotchkiss R, Opal S, Linch S, Danielson E, Kelly A, Tamakawa R, Lee J, Gold J, Castillo E, Schluns K, Jinquan T, Jing C, Jacobi H, Reimert C, Millner A, Quan S, Hansen J, Dissing S, Malling H, Skov P, Poulsen L, Driscoll K, Fisher B, Kraskauskas D, Martin E, Farkas D, Puri P, Massey H, Idowu M, Brophy D, Voelkel N, Fowler A, Natarajan R, Doi K, Leelahavanichkul A, Hu X, Sidransky K, Zhou H, Qin Y, Eisner C, Schnermann J, Yuen P, Star R, Miyaji T, Hu X, Yuen P, Muramatsu Y, Iyer S, Hewitt S, Star R, Yasuda H, Yuen P, Hu X, Zhou H, Star R, Zhang Y, Li M, Meng M, Qin C, Hotchkiss R, Swanson P, Freeman B, Tinsley K, Cobb J, Matuschak G, Buchman T, Karl I, Hotchkiss R, Tinsley K, Swanson P, Schmieg R, Hui J, Chang K, Osborne D, Freeman B, Cobb J, Buchman T, Karl I, Zingarelli B, Piraino G, Hake P, O’Connor M, Denenberg A, Fan H, Cook J. A robust scoring system to evaluate sepsis severity in an animal model. BMC Res Notes 7: 233, 2014. 67. Strawbridge AB, Blum JS. Autophagy in MHC class II antigen processing. Curr Opin Immunol 19: 87–92, 2007. 68. Takeuchi H, Kondo Y, Fujiwara K, Kanzawa T, Aoki H, Mills GB, Kondo S. Synergistic augmentation of rapamycin-induced autophagy in malignant glioma cells by phosphatidylinositol 3-kinase/protein kinase B inhibitors. Cancer Res 65: 3336–3346, 2005. 69. Thelemann C, Eren RO, Coutaz M, Brasseit J, Bouzourene H, Rosa M, Duval A, Lavanchy C, Mack V, Mueller C, Reith W, Acha-Orbea H. Interferon-γ induces expression of MHC class II on intestinal epithelial cells and protects mice from colitis. PLoS One 9: 1–10, 2014. 70. Turco F, Sarnelli G, Cirillo C, Palumbo I, De Giorgi F, D’Alessandro A, Cammarota M, Giuliano M, Cuomo R. Enteroglial-derived S100B protein integrates bacteria-induced Toll-like receptor signalling in human enteric glial cells. Gut 63: 105–15, 2014. 154 71. Vilalta A, Brown GC. Neurophagy, the phagocytosis of live neurons and synapses by glia, contributes to brain development and disease. FEBS J. (2017). doi: 10.1111/febs.14323. 72. Volchenkov R, Karlsen M, Jonsson R, Appel S. Type 1 regulatory T cells and regulatory B cells induced by tolerogenic dendritic cells. Scand J Immunol 77: 246–254, 2013. 73. Wang D, Wu F, Yuan H, Wang A, Kang GJ, Truong T, Chen L, McCallion AS, Gong X, Li S. Sox10+ Cells Contribute to Vascular Development in Multiple Organs-Brief Report. Arterioscler Thromb Vasc Biol 37: 1727–1731, 2017. 74. Wardowska A. The epigenetic face of lupus: Focus on antigen-presenting cells. Int. Immunopharmacol. 81 Elsevier B.V.: 106262, 2020. 75. Wieczorek M, Abualrous ET, Sticht J, Álvaro-Benito M, Stolzenberg S, Noé F, Freund C. Major histocompatibility complex (MHC) class I and MHC class II proteins: Conformational plasticity in antigen presentation. Front Immunol 8: 1–16, 2017. 76. Wildenberg ME, Vos ACW, Wolfkamp SCS, Duijvestein M, Verhaar AP, Te Velde AA, Van Den Brink GR, Hommes DW. Autophagy attenuates the adaptive immune response by destabilizing the immunologic synapse. Gastroenterology 142: 1493-1503.e6, 2012. 77. Wood JD, Liu S, Drossman DA, Ringel Y, Whitehead WE. Anti-enteric neuronal antibodies and the irritable bowel syndrome. J Neurogastroenterol Motil 18: 78–85, 2012. 78. Xia F, Deng C, Jiang Y, Qu Y, Deng J, Cai Z, Ding Y, Guo Z, Wang J. IL4 (interleukin 4) induces autophagy in B cells leading to exacerbated asthma. Autophagy 14: 450–464, 2018. 79. Xiao W, Wang W, Chen W, Sun L, Li X, Zhang C, Yang H. GDNF is Involved in the Barrier-Inducing Effect of Enteric Glial Cells on Intestinal Epithelial Cells Under Acute Ischemia Reperfusion Stimulation. Mol Neurobiol 50: 274–289, 2014. 80. Yang H, Plevy SE, Taylor K, Tyan D, Fischel-Ghodsian N, McElree C, Targan SR, Rotter JI. Linkage of Crohn’s disease to the major histocompatibility complex region is detected by multiple non-parametric analyses. Gut 44: 519–526, 1999. 81. Zhao J, Wang H, Yang H, Zhou Y, Tang L. Autophagy induction by rapamycin ameliorates experimental colitis and improves intestinal epithelial barrier function in IL-10 knockout mice. Int Immunopharmacol 81: 105977, 2020. 82. Zhou H, Li B, Li J, Wu T, Jin X, Yuan R, Shi P, Zhou Y, Li L, Yu F. Dysregulated T Cell Activation and Aberrant Cytokine Expression Profile in 155 Systemic Lupus Erythematosus. Mediators Inflamm 2019: 8450947, 2019. 83. Zhou M, Xu W, Wang J, Yan J, Shi Y, Zhang C, Ge W, Wu J, Du P, Chen Y. Boosting mTOR-dependent autophagy via upstream TLR4-MyD88-MAPK signalling and downstream NF-κB pathway quenches intestinal inflammation and oxidative stress injury. EBioMedicine 35: 345–360, 2018. 84. Zielinski CE, Mele F, Aschenbrenner D, Jarrossay D, Ronchi F, Gattorno M, Monticelli S, Lanzavecchia A, Sallusto F. Pathogen-induced human T H17 cells produce IFN-γ or IL-10 and are regulated by IL-1β. Nature 484: 514–518, 2012. 156 CHAPTER 4: Summary, Key Findings and Future Directions 157 Summary Enteric glia, as part of the ENS, play important roles in the maintenance of ENS health and modulation of enteric neuronal activity. Enteric glial cells express and release both pro- and anti-inflammatory mediators (1, 7, 12, 17, 18, 28), and behave similarly to atypical antigen presenting cells by expressing MHC II molecules in select conditions (2, 9). However, it has not been shown whether the expressed MHC II molecules are functional or whether enteric glial cells can carry out other antigen presenting cell functions in an in vivo system. Our study here examines enteric glial behavior in the context of an in vivo murine system with low inflammatory stress. We isolate glial contributions to overall immune and inflammatory responses by using a glial-specific inducible MHC II KO (Sox10CreERT2;IABfl/fl). Overall, our findings suggest that the enteric glia may be responding to a low dose or initial inflammatory challenge by inducing anti-inflammatory effects via autophagy-mediated MHC II expression to maintain homeostasis. Key Findings Enteric glial cells express MHC II in response to low dose IFNγ and LPS Animals challenged with a low dose of IFNγ and LPS was sufficient in eliciting an increase in MHC II expression in enteric glia. We found that the glial MHC II expression was similar in morphology to glial MHC II expressed in human Chagas disease patients (2), and this MHC II expression was in the absence of macroscopic or microscopic intestinal inflammation or tissue damage. Our results suggest that enteric glia are sensitive to the local microenvironment and may represent an ‘early responder’ to 158 inflammatory stress. Together with the findings that normal non-inflamed patients occasionally show enteric glial MHC II expression, these results suggest that the early expression of MHC II as a response to inflammatory challenge may have anti- inflammatory effects in order to maintain homeostasis. Compared to previous studies that examined enteric glial cells in the context of Chagas disease and Crohn’s disease patients, our mouse model did not experience overt activation of the inflammatory pathway and phenotypes dominated by the activation of other immune cells (2, 9). Therefore, we are able to isolate glial responses inflammatory stress and not their response to cytokine signaling from activated immune cells. Enteric glial MHC II alter T-lymphocyte activation Previously, the expression of enteric glial MHC II has not been demonstrated to be a functional molecule in modulating T-lymphocyte activation in vivo. Kermarrec et al. have shown that enteric glial MHC II have an inhibitory effect on T-lymphocyte proliferation, but the study was performed in vitro with primary cultured human enteric glial cells co-cultured with activated T-lymphocytes (16). Our results in a mouse in vivo system demonstrate that glial MHC II expression are functional, and influence T- lymphocyte levels as measured in regional mesenteric lymph nodes. In addition, we show that the Th17 and Treg subpopulations are the primary subtypes affected by enteric glial MHC II expression. Our data suggests that enteric glial MHC II may be involved in the pro-inflammatory and subsequent Treg-mediated inhibition of T-lymphocyte activation. Alternatively, MHC II expression on enteric glia may induce an anti- inflammatory phenotype in Th17 T-lymphocytes to contribute alongside Treg activities and maintain homeostasis (21, 24). Despite influencing the activation state of T- 159 lymphocytes, our data show that cytokine levels at the tissue or serum level remain unchanged, suggesting that enteric glial have more of an impact on the local microenvironment around the ENS than at the regional or whole-body level. Enteric glial MHC II expression is derived from autophagy pathways MHC II molecules are responsible for loading and presenting peptide antigens that were generally derived from the phagocytosis of extracellular molecules (26). Our results show that, despite enteric glial cells expression of MHC II molecules, they do not phagocytose extracellular antigens when challenged with IFNγ and LPS. Furthermore, our data shows that the activation of autophagy is necessary, but not sufficient to induce MHC II expression in enteric glia. Our findings suggest that enteric glia present intracellular antigens, and potentially self-antigens, for presentation onto MHC II molecules. Given that cells maintain a basal level of autophagic presentation of self- peptides onto MHC II while maintaining immune tolerance and the stimulation of autophagy ameliorates inflammation (19, 30), our data suggest that autophagy activation in enteric glia may have an anti-inflammatory effect when challenged with IFNγ and LPS. Together with our other findings, this paints a picture that enteric glia are actively attempting to maintain homeostasis in the local ENS microenvironment when faced with initial pro-inflammatory stimuli. This may help maintain proper ENS functioning prior to overt activation of pro-inflammatory immune responses that can lead to ENS damage and GI dysfunction. 160 Study Limitations Pharmacological modulators We used Rapamycin and 3-methyladenine as chemicals to stimulate or inhibit the autophagy pathway. Our findings and conclusions from experiments assume that these molecules are specific in their target activity and do not have other side effects that may confound findings. Sex of experimental animals Cytokine quantification experiments were performed in only male mice. As sex is an important variable in cytokine levels, the changes in cytokine levels observed in our studies may not be applicable to a wider population. MHC II knockout animals Our MHC II KO mouse strain is driven by the Sox10 promoter to target enteric glia in our experiments. Any cells actively utilizing the Sox10 promoter would be ablated in MHC II. We examined mesenteric lymph nodes and spleens and did not see evidence of Sox10+ cells. However, whether other Sox10+ cells in the periphery influence our findings is not known. Future Directions Determination of antigens presented on enteric glial MHC II Our findings showed that the expression of enteric glial MHC II is functional in modulating T-lymphocyte activation and is mediated by the autophagy pathway. This suggests the possibility that the peptides presented on glial MHC II are intracellular ‘self’ 161 peptides. Since the presentation of peptides on MHC II molecules can dictate the target of immune responses, it would be interesting to see what peptides enteric glial cells are presenting on their MHC II molecules. In order to determine the identity of peptides expressed to T-lymphocytes, mass spectrometry on peptides eluted from enteric glial MHC II would be beneficial. Enteric glial autophagy effects in modulating T-lymphocyte activation As discussed earlier, our results show that enteric glial MHC II expression is dependent on the activation of the autophagy pathway. Given that our data also show that enteric glial MHC II expression can influence T-lymphocyte activation, the next step would be to study the direct effects of autophagy modulation on T-lymphocyte activation, and whether a similar level of T-lymphocyte activation is seen in animals with inhibited autophagy compared to MHC II KO animals. Glial MHC II contributions in overt inflammation Our studies here were based on using a low dose of IFNγ and LPS as an inflammatory stimulus. While this inflammatory challenge is beneficial in isolating contributions from enteric glia and not from other immune cells, it would be interesting to study whether enteric glial MHC II expression have noticeable effects on the inflammatory pathway during stronger inflammatory challenge and overt inflammation. Utilizing a stronger inflammatory challenge would be beneficial in studying whether enteric glial behavior changes based on inflammation intensity. Furthermore, examining the impact of glial MHC II in stronger inflammatory stress and established inflammation 162 would give relevant findings to the pathogenesis of inflammatory bowel disease Crohn’s and ulcerative colitis. Sympathetic effects on enteric glial-mediated T-lymphocyte activation The sympathetic and parasympathetic nervous systems innervate and modulate immune activation and ENS activity (23, 25). Furthermore, the autonomic nervous system innervates enteric glial cells in the ENS (6, 10). Therefore, a future set of experiments could examine the effects autonomic activity has on enteric glial autophagy activation, expression of MHC II, and their ability to modulate T-lymphocyte activation. We performed pilot studies examining MHC II expression in the wildtype control animals after surgical sympathectomy (removal of the celiac and inferior mesenteric ganglia), chemical sympathectomy (single subcutaneous injection of 100mg/kg 6- hydroxydopamine (6-OHDA)), or norepinephrine depletion (single subcutaneous injection of 0.5mg/kg reserpine). Tissues harvested 7 days after treatments demonstrate a trend in increasing enteric glial MHC II expression in surgical sympathectomy and reserpine-treated conditions, but not in 6-OHDA-mediated chemically sympathectomized animals (Fig.4.1a,b). Since these results were observed in wildtype tissues without inflammatory challenge, changes in autonomic innervation to enteric glia in tissues and animals challenged with inflammatory stimuli may show more significant results. As mentioned above, it would be beneficial to examine how autonomic activity affects autophagy activation in enteric glia, and, by performing sympathectomy treatments to MHC II KO animals, we can elucidate the effects of autonomic activity on enteric glial activation of T-lymphocytes. 163 Figure 4.1: Autonomic innervation onto enteric glia may modulate MHC II expression. (A) Representative images of GFAP (grey) and MHC II (red) immunofluorescent labeling in enteric glia (scale bar: 50µm for all images) (B) Quantified mean fluorescent intensity of MHC II labeling on GFAP-expressing enteric glia in the myenteric plexus (n=3-5 mice; student’s unpaired t-test). 164 Local renin angiotensin effects Finally, the local renin angiotensin system within the GI tract may contribute to intestinal inflammation. Studies within the GI tract and CNS demonstrate that the binding of the prorenin receptor by prorenin or renin, along with the binding of angiotensin receptor 1 by angiotensin II can elicit pro-inflammatory effects, oxidative stress, and neuronal damage (3, 8, 31, 33) (Fig.4.2). In contrast, the binding of angiotensin receptor 2 by angiotensin II and the mas receptor by angiotensin (1-7) leads to anti-inflammatory effects, decreased oxidative stress, and increased neuronal survival (4, 11, 15, 20, 27, 29) (Fig.4.2). We performed pilot studies to examine the myenteric plexus and the expression of compounds associated with the renin- angiotensin pathway. In this collaborative pilot study with Dr. Yumei Feng Earley (University of Nevada), we examined the myenteric plexus of mice fed with a high fat diet compared to controls. Since a high fat diet in mice can lead to intestinal inflammation (5), we examined the expression of angiotensin converting enzyme-2 within the myenteric plexus as a measure of potential anti-inflammatory behavior in enteric glia. Our preliminary results show that the expression of angiotensin converting enzyme-2 is in a punctate morphology in enteric glia, and expression is increased in the high fat diet condition compared to control animals (P = .0417) (Fig.4.3a,b). These preliminary results suggest that enteric glia play a role in shifting the local renin- angiotensin pathway towards a more anti-inflammatory phenotype in the context of a high fat diet to potentially negate inflammatory effects due to elevated dietary fat. Furthermore, we will examine the role enteric neurons play in renin-induced inflammation with the use of transgenic animals with a neuron-specific KO in the 165 prorenin receptor, developed by Dr. Yumei Feng Earley (University of Nevada). These studies will help further elucidate the contributions of the local renin-angiotensin system in inflammatory stress in the GI tract. Although our preliminary study is lacking in power, our findings, like others (13, 14, 22, 32), suggest that the local renin-angiotensin system is at play in the GI tract and is involved in regulating intestinal inflammation. However, the role the ENS plays in this pathway is largely unknown and warrants further study. Taken together, the data presented in this dissertation unveils a novel role that enteric glia play as an immune modulator by interacting and controlling T-lymphocyte activation. Together with proposed future experiments, the role enteric glia play as an antigen presenting cell can be further elucidated in the context of intestinal inflammation, leading to potentially new therapeutic targets for inflammatory bowel diseases. 166 Figure 4.2: The renin-angiotensin pathway neuronal damage and inflammation. The renin-angiotensin pathway works through several receptors (prorenin receptor, angiotensin receptor 1, angiotensin receptor 2, and mas receptor) to bring about pro- and anti-inflammatory effects. Angiotensin II can bind both angiotensin receptor 1 and angiotensin receptor 2 to bring about opposing effects, but the angiotensin II molecule is generally regarded as pro-inflammatory. Angiotensin converting enzyme 2 shifts the renin-angiotensin pathway away from a pro-inflammatory phenotype to a more anti- inflammatory phenotype by increasing production of angiotensin (1-7) and binding of mas receptor. 167 Figure 4.3: Enteric glia increase expression of ACE2 during high fat diet conditions. (A) Representative Images of GFAP (grey) and ACE2 (red) immunofluorescent labeling in enteric glia. (B) Quantified mean fluorescent intensity of ACE2 labeling on GFAP-expressing enteric glia in the myenteric plexus (n=3 mice; student’s unpaired t-test; *P < .05) 168 REFERENCES 169 REFERENCES 1. Abdo H, Mahé MM, Derkinderen P, Bach-Ngohou K, Neunlist M, Lardeux B. The omega-6 fatty acid derivative 15-deoxy-Δ12,14-prostaglandin J2 is involved in neuroprotection by enteric glial cells against oxidative stress. J Physiol 590: 2739–50, 2012. 2. Barcelos Morais Da Silveira A, De Oliveira EC, Neto SG, Luquetti AO, Toshio Fujiwara R, Correa Oliveira R, Brehmer A, Barcelos A, Oliveira EC De, Neto SG, Luquetti AO, Toshio R, Correa R, Brehmer A. Enteroglial cells act as antigen-presenting cells in chagasic megacolon. Hum Pathol 42: 522–532, 2011. 3. Benicky J, Sánchez-Lemus E, Pavel J, Saavedra JM. Anti-inflammatory effects of angiotensin receptor blockers in the brain and the periphery. Cell Mol Neurobiol 29: 781–792, 2009. 4. Bennion DM, Steckelings UM, Sumners C. Neuroprotection via AT2 receptor agonists in ischemic stroke. Clin. Sci. 132 Portland Press Ltd: 1055–1067, 2018. 5. Cani PD, Bibiloni R, Knauf C, Waget A, Neyrinck AM, Delzenne NM, Burcelin R. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes 57: 1470–1481, 2008. 6. Costantini TW, Bansal V, Krzyzaniak M, Putnam JG, Peterson CY, Loomis WH, Wolf P, Baird A, Eliceiri BP, Coimbra R. Vagal nerve stimulation protects against burn-induced intestinal injury through activation of enteric glia cells. Am J Physiol Gastrointest Liver Physiol 299: G1308–G1318, 2010. 7. Fagbemi AO, Torrente F, Puleston J, Lakhoo K, James S, Murch SH. Enteric Neural Disruption in Necrotizing Enterocolitis Occurs in Association With Myenteric Glial Cell CCL20 Expression. J Pediatr Gastroenterol Nutr 57: 788–793, 2013. 8. Garg M, Royce SG, Tikellis C, Shallue C, Batu D, Velkoska E, Burrell LM, Patel SK, Beswick L, Jackson A, Britto K, Lukies M, Sluka P, Wardan H, Hirokawa Y, Tan CW, Faux M, Burgess AW, Hosking P, Monagle S, Thomas M, Gibson PR, Lubel J. Imbalance of the renin-angiotensin system may contribute to inflammation and fibrosis in IBD: A novel therapeutic target? Gut 69: 841–854, 2020. 9. Geboes K, Rutgeerts P, Ectors N, Mebis J, Penninckx F, Vantrappen G, Desmet VJ. Major histocompatibility class II expression on the small intestinal nervous system in Crohn’s disease. Gastroenterology 103: 439–447, 1992. 10. Gulbransen BD, Bains JS, Sharkey KA. Enteric Glia Are Targets of the Sympathetic Innervation of the Myenteric Plexus in the Guinea Pig Distal Colon. J 170 Protective in Lipopolysaccharide‐Induced Inflammation and Renal Injury. FASEB 11. Hussain T, Dhande I, Patel SN, Ali Q. Angiotensin AT2 Receptor Stimulation is Neurosci 30: 6801–6809, 2010. J 32: 849.8-849.8, [date unknown]. 12. Ibiza S, García-cassani B, Ribeiro H, Carvalho T, Almeida L, Marques R, Misic AM, Bartow-McKenney C, Larson DM, Pavan WJ, Eberl G, Grice EA, Veiga-Fernandes H, Eberi G, Grice EA, Veiga-Fernandes H, Eberl G, Grice EA, Veiga-Fernandes H. Glial-cell-derived neuroregulators control type 3 innate lymphoid cells and gut defence. Nature 535: 440–443, 2016. 13. Inokuchi Y, Morohashi T, Kawana I, Nagashima Y, Kihara M, Umemura S. Amelioration of 2,4,6-trinitrobenzene sulphonic acid induced colitis in angiotensinogen knockout mice. Gut 54: 349–356, 2005. 14. Jahovic N, Ercan F, Gedik N, Yüksel M, Şener G, Alican I. The effect of angiotensin-converting enzyme inhibitors on experimental colitis in rats. Regul Pept 130: 67–74, 2005. 15. Kamel AS, Abdelkader NF, Abd El-Rahman SS, Emara M, Zaki HF, Khattab MM. Stimulation of ACE2/ANG(1–7)/Mas Axis by Diminazene Ameliorates Alzheimer’s Disease in the D-Galactose-Ovariectomized Rat Model: Role of PI3K/Akt Pathway. Mol Neurobiol 55: 8188–8202, 2018. 16. Kermarrec L, Durand T, Neunlist M, Naveilhan P, Neveu I. Enteric glial cells have specific immunosuppressive properties. J Neuroimmunol 295–296: 79–83, 2016. 17. Liñán-Rico A, Turco F, Ochoa-Cortes F, Harzman A, Needleman BJ, Arsenescu R, Abdel-Rasoul M, Fadda P, Grants I, Whitaker E, Cuomo R, Christofi FL. Molecular Signaling and Dysfunction of the Human Reactive Enteric Glial Cell Phenotype: Implications for GI Infection, IBD, POI, Neurological, Motility, and GI Disorders. Inflamm Bowel Dis 22: 1812–34, 2016. 18. Le Loupp A-G, Bach-Ngohou K, Bourreille A, Boudin H, Rolli-Derkinderen M, Denis MG, Neunlist M, Masson D. Activation of the prostaglandin D2 metabolic pathway in Crohn’s disease: involvement of the enteric nervous system. BMC Gastroenterol 15: 112, 2015. 19. Macias-Ceja DC, Cosín-Roger J, Ortiz-Masiá D, Salvador P, Hernández C, Esplugues J V, Calatayud S, Barrachina MD. Stimulation of autophagy prevents intestinal mucosal inflammation and ameliorates murine colitis. Br J Pharmacol 174: 2501–2511, 2017. 20. Matavelli LC, Siragy HM. AT2 receptor activities and pathophysiological implications. J Cardiovasc Pharmacol 65: 226–232, 2015. 171 21. McGeachy MJ, Bak-Jensen KS, Chen Y, Tato CM, Blumenschein W, McClanahan T, Cua DJ. TGF-β and IL-6 drive the production of IL-17 and IL-10 by T cells and restrain TH-17 cell-mediated pathology. Nat Immunol 8: 1390– 1397, 2007. 22. Mizushima T, Sasaki M, Ando T, Wada T, Tanaka M, Okamoto Y, Ebi M, Hirata Y, Murakami K, Mizoshita T, Shimura T, Kubota E, Ogasawara N, Tanida S, Kataoka H, Kamiya T, Alexander JS, Joh T. Blockage of angiotensin II type 1 receptor regulates TNF-α-induced MAdCAM-1 expression via inhibition of NF-κB translocation to the nucleus and ameliorates colitis. Am J Physiol - Gastrointest Liver Physiol 298, 2010. 23. Mourad FH, Saadé NE. Neural regulation of intestinal nutrient absorption. Prog Neurobiol 95: 149–162, 2011. 24. Musuraca G, De Matteis S, Napolitano R, Papayannidis C, Guadagnuolo V, Fabbri F, Cangini D, Ceccolini M, Giannini MB, Lucchesi A, Ronconi S, Mariotti P, Savini P, Tani M, Fattori PP, Guidoboni M, Martinelli G, Zoli W, Amadori D, Carloni S. IL-17/IL-10 double-producing T cells: New link between infections, immunosuppression and acute myeloid leukemia. J Transl Med 13: 1– 10, 2015. 25. De Ponti F, Giaroni C, Cosentino M, Lecchini S, Frigo G. Adrenergic mechanisms in the control of gastrointestinal motility: from basic science to clinical applications. Pharmacol Ther 69: 59–78, 1996. 26. Roche PA, Furuta K. The ins and outs of MHC class II-mediated antigen processing and presentation. Nat Rev Immunol 15: 203–16, 2015. 27. Rodrigues Prestes TR, Rocha NP, Miranda AS, Teixeira AL, Simoes-e-Silva AC. The Anti-Inflammatory Potential of ACE2/Angiotensin-(1-7)/Mas Receptor Axis: Evidence from Basic and Clinical Research. Curr Drug Targets 18, 2017. 28. Rosenbaum C, Schick MA, Wollborn J, Heider A, Scholz C-J, Cecil A, Niesler B, Hirrlinger J, Walles H, Metzger M. Activation of Myenteric Glia during Acute Inflammation In Vitro and In Vivo. PLoS One 11: e0151335, 2016. 29. Sabuhi R, Ali Q, Asghar M, Al-Zamily NRH, Hussain T. Role of the angiotensin II AT 2 receptor in inflammation and oxidative stress: opposing effects in lean and obese Zucker rats. Am J Physiol Physiol 300: F700–F706, 2011. 30. Schmid D, Pypaert M, Münz C. Antigen-loading compartments for major histocompatibility complex class II molecules continuously receive input from autophagosomes. Immunity 26: 79–92, 2007. 31. Shi P, Grobe JL, Desland FA, Zhou G, Shen XZ, Shan Z, Liu M, Raizada MK, Sumners C. Direct pro-inflammatory effects of prorenin on microglia. PLoS One 9: 92937, 2014. 172 32. Spencer AU, Yang H, Haxhija EQ, Wildhaber BE, Greenson JK, Teitelbaum DH. Reduced severity of a mouse colitis model with angiotensin converting enzyme inhibition. Dig Dis Sci 52: 1060–1070, 2007. 33. Worker CJ, Li W, Feng CY, Souza LAC, Gayban AJB, Cooper SG, Afrin S, Romanick S, Ferguson BS, Earley YF. The neuronal (pro)renin receptor and astrocyte inflammation in the central regulation of blood pressure and blood glucose in mice fed a high-fat diet. Am J Physiol - Endocrinol Metab 318: E765– E778, 2020. 173