.. x; n akin x:;a.a. I. z .2 Us! :1 S g, Bali: .5 . :2; 8.; a. £135” ‘ 492... ...S..nur .05.. J}... $9.13!.- .I ). IEoJLEIII-Y .03! 3-]: I....|\..‘.}l§ [2141:3’ SPEC!!! b). rI....l.>O..~I.(.w t...) £1435..l’. _ . : 3.3 $21. .A. It: :: t A . .. zwwfiiufl l IIHHAr-s :3] j EdJnState L University This is to certify that the dissertation entitled The in vivo characterization of spleen dendritic cell populations of vitamin A-deficient C 57BL/6J mice presented by David Michael Duriancik has been accepted towards fulfillment of the requirements for the Doctoral degree in Human Nutrition W ZW Major Professor's Signature 02/00/020“) Date MSU is an Affirmative Action/Equal Opportunity Employer PLACE IN RETURN BOX to remove this checkout from your record. To AVOID FINES return on or before date due. MAY BE RECALLED with earlier due date if requested. DATE DUE DATE DUE DATE DUE 5/08 K:IProi/Aoc&Pres/ClRC/Dateoue.indd THE [N V1 V0 CHARACTERIZATION OF SPLEEN DENDRITIC CELL POPULATIONS OF VITAMIN A-DEFICIENT C57BL/6J MICE By David Michael Duriancik A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Human Nutrition 2010 ABSTRACT The in vivo characterization of spleen dendritic cell populations of vitamin A-deficient C57BL/6J mice By David Michael Duriancik Vitamin A deficiency affects 125 million preschool children each year. Vitamin A-deficient populations have decreased T—dependent antibody responses and unaffected or increased cell-mediated immune responses. The impaired T-dependent antibody response is due to decreased stimulation by T helper (Th) 2 cells as well as cross- regulation by increased Th1 cells. In the mouse, CD1 lb+ myeloid dendritic cells (DCs) stimulate Th2 responses while CD80? lymphoid DCs stimulate Th1 responses. In addition, vitamin A is important for myelopoiesis and differentiation of myeloid progenitors into dendritic cells (DCs) and neutrophils. Therefore, we hypothesized that vitamin A-deficient mice would have decreased numbers of myeloid DCs and increased numbers of neutrophils compared to vitamin A-sufficient mice. We developed a multicolor flow cytometry protocol for identifying DC populations from individual C57BL/6J mouse spleen. The protocol is novel, lacking any enrichment procedures or DC proliferating conditions. Previously, researchers have used various enrichment procedures, pooling of tissues from multiple animals, or stimulating DC proliferation to obtain sufficient numbers of cells to analyze. These procedures potentially skew analysis of DCs to select DC populations or increase variability. Particularly when studying vitamin A deficiency, it is important to analyze animals individually. Each animal has variable vitamin A status, other health parameters, and unique developmental age. Using the protocol we developed, we show that vitamin A-deficient animals have increased lymphoid DCs (R2 0.41, liver RAE B -0.63), lymphoid to myeloid DC ratio (R2 0.52, liver RAE [3 -0.68), neutrophils (R2 0.20, liver RAE B -0.44), memory CD8+ T lymphocytes (R2 0.38, liver RAE is -0.62), and CD4+ T lymphocytes (R2 0.36, liver RAE B 0.62) in their spleen. The increased lymphoid DC percentage was unexpected, but may be inducing memory CD8+ T cells to proliferate and produce interferon- (IFN) 7. The cytokine environment is therefore Th1 and memory CD8+ T cell biased and the increased Th1 environment would be expected to down-regulate the Th2 antibody-mediated immune response. Therefore, the slight decrease in myeloid DCs (R2 0.22, liver RAE B 0.32) combined with the increase in lymphoid DCs and memory CD8+ T cells provides mechanistic evidence for the depressed antibody responses observed in vitamin A- deficient populations. The data we present here confirms reports of others, provides novel evidence of the role of vitamin A on DC homeostasis, and provides a comprehensive overview of the immune system in vitamin A-deficient mice. We confirm that vitamin A does not affect splenic B lymphocyte (R2 0.08, liver RAE [3 -0.00) and CD8+ T lymphocyte numbers (R2 0.10, liver RAE B -0.07), and show that vitamin A deficiency also does not alter precursor DC (R2 0.09, liver RAE [3 0. l 0) or plasmacytoid DC numbers (R2 0.02, liver RAE B 0.08). We show that the immune response bias previously reported in vitamin A deficiency correlates with an increased ratio of lymphoid to myeloid DCs in vitamin A- deficient animals. We confirm neutrophils are increased in vitamin A-deficient populations, but not to the dramatic extent previously reported. ACKNOWLEDGMENTS I would like to show my appreciation for the support and advice of my committee members and the Department of Food Science and Human Nutrition. I am grateful for the opportunity to have worked closely with my committee members and other faculty members. This project could not have been completed without each of their individual expertise. Dr. Zile provided expertise in vitamin A, Dr. Davis provided statistical expertise, Dr. Gerlach provide expertise in immunology, and Dr. Bennink provided expertise in diet composition and animal welfare. I would also like to acknowledge Dr. Louis King, director of the Michigan State University flow cytometry core facility, for his help in developing and analyzing the flow cytometry data. I appreciate the assistance of other students in Dr. Hoag’s laboratory. Denise Lackey has been a tremendous help through assisting with experiments, interpreting data, and answering questions. Carmen Yu and Shanna Ashley assisted with experiments. All of Dr. Hoag’s former students created a lively learning environment and my time spent in the laboratory was an enjoyable experience. I would also like to thank my family, friends, and colleagues for their support throughout my graduate experience. Most of all I would like to thank my advisor, Dr. Hoag. In every step of the graduate education process, she has been a valuable mentor and friend. I learned so much from working with Dr. Hoag. She took a chance by accepting me into her laboratory and I hope to make her proud throughout my career. Dr. Hoag, I sincerely thank you for all that you have done for me. TABLE OF CONTENTS List of tables ...................................................................................................................... vii List of figures ................................................................................................................... viii Introduction .......................................................................................................................... 1 Chapter 1: Literature review ................................................................................................ 3 Immune system overview .............................................................................................. 3 Dendritic cells ................................................................................................................ 5 Vitamin A metabolism ................................................................................................... 7 Vitamin A function ...................................................................................................... 10 Vitamin A deficiency ................................................................................................... 13 Vitamin A and innate immunity .................................................................................. 14 Vitamin A and adaptive immunity ............................................................................... 17 Vitamin A and DCs ...................................................................................................... 19 Experiment rationale .................................................................................................... 21 Chapter 2: The identification and enumeration of dendritic cell populations from individual mouse spleen and Peyer’s patches using flow cytometric analysis .................. 24 Abstract ........................................................................................................................ 25 Introduction .................................................................................................................. 26 Methods ........................................................................................................................ 27 Animals .................................................................................................................. 27 Tissue isolation ...................................................................................................... 28 Antibody staining ................................................................................................... 29 F low cytometry acquisition & analysis .................................................................. 29 Flow cytometric analysis gating strategies ............................................................ 30 Population calculations and statistical analysis ..................................................... 32 Results .......................................................................................................................... 32 Animal characteristics ............................................................................................ 32 Average immune cell numbers obtained from healthy C57BL/6J mouse spleen and Peyer’s patches ............................................................................................................ 33 Discussion .................................................................................................................... 35 Acknowledgements ...................................................................................................... 40 Chapter 3: Vitamin A deficiency alters splenic dendritic cell subsets and increases CD8+Gr-1+ memory T lymphocytes in C57BL/6J mice. ................................................. 57 Abstract ........................................................................................................................ 58 Introduction .................................................................................................................. 59 Methods ........................................................................................................................ 61 Animals .................................................................................................................. 61 Vitamin A analysis ................................................................................................. 61 Tissue processing ................................................................................................... 62 Flow cytometry ...................................................................................................... 62 Statistical analysis .................................................................................................. 63 Results .......................................................................................................................... 63 Discussion .................................................................................................................... 66 Chapter 4: Conclusions & future directions ..................................................................... 101 Flow cytometry of DCs .............................................................................................. 101 Murine model of vitamin A deficiency ...................................................................... 101 Future directions ........................................................................................................ 103 Appendix I ....................................................................................................................... 108 Appendix II ...................................................................................................................... 1 18 Literature cited ................................................................................................................. 122 vi LIST OF TABLES Chapter 2 Table 2.1. Antibody-fluorochrome conjugate reagents employed in labeling cell suspensions for flow cytometric analysis. ....................................................... 49 Table 2.2. Antigen expression of immune cell populations in mouse spleen and Peyer's patches used in gating strategy. ....................................................................... 50 Table 2.3. Physiological characteristics of C57BL/6J mice used in the studies. .............. 52 Table 2.4. Percentage and total numbers of dendritic cell populations of C57BL/6J spleen and Peyer's patches. .............................................................................. 53 Table 2.5. Percentage and total cell numbers of non-dendritic immune cell populations enumerated in C57BL/6J spleen and Peyer's patches. ..................................... 55 Chapter 3' Table 3.1. Liver weight of VAS and VAD male and female animals. ............................. 69 Table 3.2. Spleen weight of VAS and VAD male and female animals. ........................... 70 Appendix 1 Supplemental Table 1.1. BD® Biosciences LSR II flow cytometer laser parameters used in flow cytometry data collection. ................................................................. l 15 Supplemental Table 1.2. BD® Biosciences LSR 11 general compensation matrix. ........ l 16 vii LIST OF FIGURES Some figures in this dissertation are presented in color. Chapter 2 Figure 2.1. Gating strategy to identify dendritic cells and other immune cell populations in mouse spleen. ............................................................................................. 41 Figure 2.2. Gating strategy to identify dendritic cells and other immune cell populations in mouse Peyer's patches. ............................................................................... 45 Chapter 3 Figure 3.1. Body weights ofC57BL/6J mice consuming VAS and VAD diet. ............... 71 Figure 3.2. Vitamin A status of C57BL/6J mice. ............................................................. 73 Figure 3.3. Total spleen cells per animal of C57BL/6J mice ............................................ 77 Figure 3.4. Effects of depleted liver RAE on spleen DC populations. ............................. 79 Figure 3.5. Effects of depleted liver RAE on spleen lymphocyte populations. ................ 89 Figure 3.6. Effects of depleted liver RAE on spleen PMN ............................................... 99 Appendix I Supplemental Figure 1.1. LSR II optics block and filter scheme of the fluorescent channels utilized for data collection. ............................................................... 109 Supplemental Figure 1.2. Comparison of compensated and uncompensated data. ......... 111 Supplemental Figure 1.3. Flow diagram of Boolean logic used in sequential gating of spleen cell populations. ................................................................................. l 13 Appendix 11 Supplemental Figure 2.1. Immune cell percentages of vitamin A-deficient mice ........... 1 18 viii KEY TO SYMBOLS & ABBREVIATIONS Allophycocyanin (APC) American Institute of Nutrition (AIN) Analysis of variance (ANOVA) Antigen presenting cell (APC) Cluster of differentiation (CD) Cyan (Cy) Cytotoxic T lymphocyte (CTL) Dendritic cell (DC) F luorescein isothiocyanate (F ITC ) Fluorescence activated cell sorting (FACS) Forward scatter (FSC) Granulocyte/macrophage-colony stimulating factor (GM—CSF) Hank’s balanced salts solution (HBSS) High performance liquid chromatography (HPLC) Immunoglobulin (1g) Interferon (IFN) Interleukin (IL) Major histocompatibility complex (MHC) Matrix metalloproteinasc (MMP) Michigan State University (MSU) Monoclonal antibody (mAb) Monocyte-derived dendritic cell (mDC) Natural killer (N K) Negative (NEG) Not Applicable (N/A) Peridinin chlorophyll protein (PerCP) Peyer’s patch (PP) Phycoerythrin (PE) Polymorphonuclear neutrOphil (PMN) Positive (POS) Precursor DC (preDC) Promyelocytic leukemia (PML) Protein—energy malnutrition (PEM) Retinoic acid receptor (RAR) Retinoic acid response element (RARE) Retinoid X receptor (RXR) Retinol activity equivalents (RAE) Retinol binding protein (RBP) Side scatter (SSC) T helper (Th) Transthyretin (TTR) T regulatory (Treg) Tumor necrosis factor (TN F) Vitamin A-deficient (VAD) Vitamin A-sufficient (VAS) INTRODUCTION Each year approximately 125 million children suffer from vitamin A deficiency. Vitamin A deficiency increases the risk of infection as well as the morbidity and mortality of infection. The immune response of vitamin A-deficient populations is biased to T helper 1 (Th1) cell-mediated responses with decreased Th2 antibody responses. Previous research by others has focused on lymphocyte number and function of vitamin A-deficient populations, but vitamin A has important roles in hematopoiesis, particularly myelopoiesis. Acute promyelocytic leukemia results from a fusion of retinoic acid receptor (RAR) or to the PML gene leading to an increase in immature neutrophil numbers. In addition, previously our laboratory demonstrated that murine bone marrow cells cultured in media containing granulocyte-macrophage colony stimulating factor (GM-CSF) and retinoic acid develop into dendritic cells, while bone marrow cells cultured with depleted retinoic acid or retinoic acid antagonists develop into neutrophils. Furthermore, others have shown systemic myeloid cell, specifically neutrophil, expansion in mice depleted of vitamin A. Dendritic cells (DCs) are bone marrow derived antigen presenting cells responsible for the stimulation of naive T cells in response to infection. In the mouse, CD1 1b+ myeloid DCs stimulate Th2 immune responses, while CD80t+ lymphoid DCs stimulate Th1 immune responses. Plasmacytoid DCs secrete large amounts of type 1 intereferons (IFN), IFN-or/B. Myeloid, lymphoid and plasmacytoid DCs are mature DCs that arise from one common precursor cell circulating in blood and resident in secondary lymphoid tissues. DCs are sentinels of the body, resident in sparse numbers in every tissue, scanning the body for danger signals. Immature DCs are phagocytic cells, sampling antigens for danger signals. Upon recognition of a danger signal, DCs undergo maturation through the upregulation of antigen-presenting, T cell costimulatory, and homing receptors to stimulate naive T cells in secondary lymphoid tissues. The impaired Th2 immune responses of vitamin A-deficient populations may be the result of skewed DC subsets. In addition, the requirement of retinoic acid for DC development from bone marrow progenitors and the expansion of neutrophils of vitamin A-deficient mice provide convincing evidence of skewed DC subsets of vitamin A- deficient mice. Therefore, it is hypothesized that myeloid DCs would be decreased and neutrophils would be increased in the spleens of vitamin A-deficient mice. Multicolor flow cytometry was used to identify and compare the DC, neutrophil, and lymphocyte populations of mice with varying levels of liver stores of vitamin A. CHAPTER 1: LITERATURE REVIEW Immune System Overview The immune system is comprised of innate and adaptive arms that protect the body from infection. The innate arm consists of non-specific, ubiquitous, and immediate protection from pathogens. The adaptive arm consists of specific, localized, and delayed protection against “danger” signals. “Danger” signals are foreign substances such as pathogenic microorganisms and viruses, but also consist of altered “self” signals such as tumor cells. The innate and adaptive arms of the immune system communicate through various signals to provide the most effective protection from infection (1). The innate immune system consists of epithelial barriers, non-specific phagocytic and lytic cells, and other soluble and circulating biological factors. The epithelial barriers include both skin and mucosal tissue. Phagocytic and lytic cells include macrophages, neutrophils, and natural killer (N K) cells. Other biological factors include protease enzymes, salts, pH differences, and other proteins such as complement. These biological factors inhibit microbe proliferation or directly induce microbial cell death. Biological factors work in concert with barriers and cells to provide optimal protection, such as the antimicrobial products present in mucus. Bridging the innate and adaptive arms are the phagocytic cells. The phagocytic cells, particularly dendritic cells, take up microbes, digest them, and then present antigens to T lymphocytes. Presentation of antigen leads to the initiation of an adaptive immune response through the production and secretion of cytokines creating an environment for the most effective response to a given pathogen. Other antigen—presenting cells (APCs) in addition to dendritic cells include macrophages and B lymphocytes (1). B and T lymphocytes are cells of the adaptive immune response that recirculate through secondary lymphoid tissues, but are capable of homing to sites of infection following stimulation by an APC. The processes of antigen uptake and presentation as well as cell proliferation and differentiation that occur in an initial adaptive immune response lead to a delay in the B and T cell responses. However, previously encountered pathogens have more rapid subsequent responses due to the development of memory B and T cells. B lymphocytes produce and secrete antibody to various antigens present on a microbial cell or soluble microbial products such as a toxins. There are numerous subsets of T lymphocytes distinguished by their T cell co-receptor expression and/or function. Cluster of differentiation (CD) 4+ T cells recognize peptide antigens presented in major histocompatibility complex (MHC) - 11 proteins on APCs and develop T helper (Th) responses. There are 2 classes of Th effector cell responses; Th1 which participate in cell-mediated immunity, and Th2 which aid B lymphocyte antibody immunity. T cells expressing the CD8 T cell co-receptor, also called cytotoxic T lymphocytes, recognize antigen in the context of MHC-I proteins and receive help from a CD4+ Th1 cell to accomplish a cell-mediated response. T cells expressing the CD4 T cell co-receptor are also responsible for inducing tolerance to non-dangerous or self antigens, and this subset ofT lymphocytes is called T regulatory (Treg) cells. Thus, the adaptive immune system is a highly complex and coordinated multi—cellular response designed to defend against specific danger signals while at the same time being tolerant to self antigens (1). Cooperation between the innate and adaptive arms of the immune system is critical for optimal immune responses to danger signals as well as maintaining tolerance to self signals. APCs are the critical link between the innate and adaptive immune responses through non-specific phagocytosis of antigens, antigen processing and presentation, and cytokine secretion directing the subsequent adaptive immune response. Antigen presentation and cytokine secretion communicate the most effective T lymphocyte response to a specific pathogen that has evaded the innate immune system. Due to the cooperation and coordination of signals, any perturbation in the system can lead to malfunctions of the other components of the immune system. Dendritic Cells Dendritic cells (DCs) are hematopoietic-derived APCs that efficiently stimulate naive T lymphocytes (2-4). DCs migrate from the bone marrow and reside in peripheral tissues in an immature state (5). Under stead-state conditions, a reservoir of precursor and mature DCs are maintained in secondary lymphoid tissues, but DCs are sparse in other tissues where they function as sentinels (5, 6). DCs comprise about 1-3% of the cells of the spleen (7). Upon phagocytosis of pathogen molecules in tissues, the DCs mature and migrate to secondary lymphoid tissues, such as the spleen and lymph nodes, to stimulate T cells and induce an adaptive immune response. Maturation of DCs involves upregulation of homing receptors and other surface proteins necessary to efficiently stimulate T cells, including CD40, CD80, and CD86 (8, 9). I Various DC subpopulations exist, and each has a bias to stimulate a particular adaptive immune response. In the mouse, myeloid DCs preferentially stimulate antibody-mediated (Th2) responses, while lymphoid DCs stimulate cell-mediated (Th1) responses (10, 1 1). Plasmacytoid DCs produce large amounts of type 1 interferons (IFN), IF N-a and —B, to augment antiviral immune responses (12-15). Myeloid DC 3 express CD1 1c, MHC-II, and CD1 1b, but do not express CD80t. Lymphoid DCs express CD1 1c, MHC-II, and CD80L, but do not express CD1 1b (10, l l). Plasmacytoid DCs express CD11c, MHC-II, Gr-l , and B220 (CD45RB) (12, 16). Although controversial at one point, it is now known that these DC subpopulations transcribe and translate the various surface protein markers and do not acquire the surface proteins through cell-cell contact interactions with other cell types, such as CD8 T lymphocytes (17, 18). The respective DCs present antigens to Th cells and secrete cytokines to stimulate the appropriate adaptive immune response. DCs arise from both common myeloid or common lymphoid progenitors in the bone marrow (4, 19). Precursor DCs (preDCs) migrate from the bone marrow to the bloodstream and reside in peripheral tissues in an immature state (20-22). Upon recognition of antigens through pattern recognition receptors, such as toll-like receptors, the preDCs mature and home to secondary lymphoid tissues to stimulate naive T cells (23). Myeloid and lymphoid DCs have half-lives of 1.5 to about 3 days, while plasmacytoid DCs have a half-life of about 9 days (19, 24-26). During an infection, cytokines stimulate DC differentiation of bone marrow progenitors, DC turnover (proliferation and apoptosis), and migration from sites of infection through the lymphatic system to the draining lymph node or through blood vasculature to the spleen during a systemic infection. Humans have a slightly different repertoire of DCs compared to mice, but mouse models can be extrapolated to the human immune system (8). Human DCs may arise from common myeloid progenitors, common lymphoid progenitors, or differentiate from other cell types including blood monocytes (27). Human DCs are classified into mDC-l, mDC-2, and plasmacytoid DCs. Human plasmacytoid DCs are similar to mouse plasmacytoid DCs (15). Human mDC-l preferentially stimulate Th1 responses similar to mouse lymphoid DCs, while human mDC-2 stimulate Th2 responses similar to mouse myeloid DCs (28). Human DC research has been limited due to available tissues for analysis, and most research has been extrapolated from in vitro differentiation of immature DCs and monocytes obtained from the blood. In both humans and mice, DCs possess a high level of marker expression and functional plasticity. The DC plasticity, low percentages in a particular tissue, and lack of clear maturation processes make DC research difficult and complex. Vitamin A Metabolism Vitamin A is a generic term that encompasses retinoid and carotenoid compounds that supply humans with a biological need. The best characterized physiological forms of vitamin A are all-trans-retinoic acid, the carboxylic acid form, retinal, the aldehyde form, and retinol, the alcohol form, depending on specific physiological function and tissue (29). Mammals can not synthesize vitamin A de novo. Vitamin A is a fat-soluble vitamin obtained by humans in two different dietary forms, i.e. retinyl esters or pro- vitamin A compounds. The physiological form of vitamin A circulating in the bloodstream, retinol, can he obtained in association with the fat of animal products as retinyl esters, or retinol esterified to fatty acids. Pro-vitamin A compounds can be consumed from yellow, orange, and some dark green plants. Pro-vitamin A compounds, including or- and B-carotenes and B-cryptoxanthin, can be metabolized to retinoids through enzymatic cleavage. The enzyme 15, 15’—monooxygenase is expressed in a multitude of human tissues and converts pro-vitamin A compounds to biologically active forms of vitamin A (30). The tissues expressing 15, 15’-monooxygenase include cells of the gastrointestinal tract, reproductive organs, skin, liver, and skeletal muscle (30). The highest expression of 15,15’-monooxygenase is in the gastrointestinal tract. Retinoids, and a fraction of consumed carotenoids, are absorbed in the small intestine. Retinoid absorption is described below, and carotenoids are absorbed intact via passive diffusion and associate directly with chylomicrons (31). Retinyl esters are hydrolyzed to retinol in the gastrointestinal tract by the pancreatic and enterocyte enzymes retinyl ester hydrolases and non—specific lipases (32- 34). Retinol is absorbed by the enterocyte and esterified to a fatty acid, normally palmitate or stearate, by the enterocyte enzymes lecithinzretinol acyltransferase and acyl- CoAzretinol acyltransferase (35, 36). The retinyl ester is then complexed with cholesterol, phospholipids, other fatty acids, and binding proteins and excreted from the enterocyte into the lymphatic system within a chylomicron (37). The chylomicron circulates through the lymphatic and vascular system delivering retinyl esters and other chylomicron components to peripheral tissues (38). The depleted chylomicron, now a chylomicron remnant, will deposit the remaining retinyl esters into liver stellate cells for storage (39). Upon demand, liver reserves of retinyl esters are hydrolyzed to retinol which binds to retinol binding protein (RBP). Retinol-RBP complex next binds to transthyretin (TTR) in the liver prior to release into the bloodstream to maintain a relatively constant level of circulating retinol (35). The circulating retinol-RBP-TTR complex, in a l :1 :1 ratio, increases molecular mass and decreases loss through glomerular filtration (40-42). In a healthy state, vitamin A is circulating in the blood at a consistent level of retinol:RBP:TTR at approximately 1-2 micromolar (11M) concentration, but during acute infections the concentration may decrease (43). The membrane receptor Stra6 binds RBP and mediates the uptake of retinol into the cell, but RBP remains extracellular (44). Extracellular RBP returns to circulation and is filtered through the kidney glomerulus. A cell can store limited amounts of retinol complexed with cellular retinol binding protein or will oxidize retinol to retinal, a reversible reaction catalyzed by the enzyme retinol dehydrogenase. The retinal can then be irreversibly oxidized to retinoic acid by retinal dehydrogenase (45). The cell can store limited amounts of retinoic acid complexed with cellular retinoic acid binding protein (46). In addition, a cell can also synthesize retinyl esters as well as metabolize retinoids to polar derivatives to increase cellular reserves of retinoids (35). Vitamin A is a hydrophobic molecule. Therefore, vitamin A is soluble in fat and requires a protein carrier to be soluble in the bloodstream or cell cytosol. Liver is the main storage site of vitamin A, but adipose and other tissues can store limited amounts of vitamin A (47). Adipose and other tissues storing vitamin A can liberate the stores in a mechanism similar to that in liver (48). Retinoic acid can be carried in the bloodstream and delivered to peripheral tissues by albumin (3 8). However, adipose storage and albumin delivery of retinoic acid are believed to be minor contributions to the overall metabolism and transport of vitamin A. Vitamin A is excreted in both the feces and urine (49, 50). Vitamin A can be serially oxidized to increasingly polar metabolites; from retinyl-esters to retinyl- glucoronides (51). All-trans-retinyl B-glucuronide and other polar metabolites are secreted into bile and, if not reabsorbed, excreted in the feces (49). The glomerulus of the kidney filters apo-RBP, but holo-RBP complexed with TTR is retained due to the size of the complex, recycling retinol-protein complex back to circulation (52). However, the kidney also expresses RBP and TTR binding proteins, specifically megalin, to aid in the reabsorption of vitamin A (53). Vitamin A, in various forms, is removed from the body through biliary excretion and urinary filtration. Vitamin A Function There are three general physiological functions of vitamin A requiring different structural forms. Retinol and all-trans-retinoic acid are required for reproductive processes in both males and females (54). Retinal, specifically 11-cis-retinal, is critical in vision (55). All-trans-retinoic acid in complex with its nuclear receptor functions as a steroid hormone family transcription factor (56, 57). All-trans-retinoic acid is a ligand for a family transcription factors involved in the regulation of cell differentiation, proliferation, maturation and apoptosis. The function of all-trans-retinoic acid as a ligand for transcription factors has been extensively reviewed and will be explained in greater detail below (58-60). High intakes of vitamin A during pregnancy are teratogenic, but vitamin A, specifically retinol and all-trans-retinoic acid, are critical in reproduction (61-63). The role of all-trans-retinoic acid in reproduction is through gene regulation via nuclear receptors. However, retinol may have unique functions in reproduction. All-trans- retinoic acid alone can not support normal reproduction, supplemental retinyl esters or all-trans-retinol are required to prevent fetal resorption (64, 65). The specific role of retinol in late gestation is not clear, but is required for normal heart, brain, and eye development. The temporal-spatial distribution of vitamin A metabolites is critical in 10 reproduction to allow for all stages of fetal development and prevention of fetal resorption (54, 63). Rhodopsin, a light sensing protein of the rod cells in the eye, requires ll-cis- retinal as a cofactor (66). Ultraviolet light causes 1 1-cis-retinal to isomerize to all-trans- retinal, leading to a conformational change in the rhodopsin protein (67). The conformational change in rhodopsin signals light detection to the brain (68). All-trans- retinal can be recycled and converted back to 1 l-cis-retinal in the eye (69). In the retina, ll-cis-retinal functions in light adaptation. In the cone cells, ll-cis-retinal functions in color vision (70). The function of 1 l-cis retinal is restricted to the eye. All-trans-retinoic acid and 9-cis-retinoic acid are both ligands for the transcription factor family retinoic acid receptors (RARs). However, only 9-cis-retinoic acid has been characterized as the ligand for retinoid X receptors (RXRs). There are three isoforms of RAR (or, B, y) and RXR (or, [3, y) (59). All-trans-retinoic acid translocates from the cytosol to the nucleus and binds to RARs (59). Upon ligand binding, RARs heterodimerize with RXRS and bind to a retinoic acid response element (RARE) of target genes for regulatory function (59). The RAR/RXR heterodimer will recruit other gene promoting or suppressing transcription factors to regulate the transcription of the candidate gene (59). More than 532 genes have been described as responsive to retinoic acid (71). However, only about 30% of these genes have promoters possessing classic RAREs. Classical RAREs are six nucleotide base pairs of guanine and cytosine direct repeats separated by one, two, or five nucleotides (59, 72, 73). However, non-classical RAREs exist in which guanine and cytosine rich half-site regions are separated by any number of base pairs (59). RAR/RXR transcriptional regulation is a complex and 11 multistep process of ligand binding, translocation to RARE sites, and association with other transcription factors. Retinoid-mediated gene transcriptional control in the absence of a classic RARE is commonly documented, but the molecular mechanism(s) involved is largely unknown (59). The RXR proteins may heterodimerize with other transcription factor partners or may also homodimerize (59). In vitro, 9-cis-retinoic acid will bind to RXRs leading to homodimerization and transcriptional regulation. However, 9-cis-retinoic acid has not been measurable in tissue samples, and therefore the physiological relevance of 9-cis- retinoic acid and RXR homodimer transcriptional regulation remains controversial. RXRs also heterodimerize with other nuclear receptor superfamily transcription factor partners, including vitamin D receptor, peroxisome proliferator activation receptors, liver X receptors, and others (59). The alternate dimerization partners lead to binding to different response elements and modulation of other genes regulated at the transcriptional level. In the immune system, the role of vitamin A is generally confined to a steroid hormone family transcription factor through all-trans-retinoic acid-RAR-RXR gene regulation. Acute promyelocytic leukemia (PML) is a hematopoietic cell cancer in which the RARor gene on chromosome 15 reciprocally translocates and fuses to the PML gene on chromosome 17 (74). The fusion of RARa to PML blocks the normal maturation of granulocytes, including neutrophils. Prescription of high dose all-trans-retinoic acid has been an effective treatment option through the induction of differentiation of the cancer cells into mature granulocytes which then have a finite half-life (75). Therefore, at the 12 transcriptional level, all—trans-retinoic acid is well-established to be important in the differentiation of myeloid progenitors in the bone marrow. Vitamin A Deficiency Vitamin A deficiency is a global nutrient concern affecting approximately 125 million children each year (76). In the US, the prevalence of vitamin A deficiency estimated from National Health and Nutrition Examination Survey data may be as high as 25% depending on the cutoff of vitamin A deficiency and the subpopulation of interest (77). Incidence of vitamin A deficiency is consistently higher in children, pregnant women, and patients with fat malabsorption diseases. Vitamin A deficiency can result from inadequate intakes of vitamin A rich foods or poor absorption of fat-soluble molecules. Numerous strategies have been employed and studied to ameliorate vitamin A deficiency including supplementation programs, crop distribution, and bioengineering of crops to contain pro—vitamin A compounds, specifically B-carotene (78, 79). Vitamin A supplementation efforts are a cost effective means to improve health of populations, especially of young children. Supplementing vitamin A costs cents per dose and can reduce childhood mortality on average by 30% (80, 81). Despite these efforts, to date vitamin A deficiency remains a factor in childhood morbidity and mortality around the world. Detection of vitamin A deficiency is difficult. The high liver stores and constant release of vitamin A maintains circulating serum retinol constant at about l-2uM until chronic undemutrition depletes liver stores. Serum retinol can also be decreased during the acute phase response of an infection (43). Therefore, a lower serum retinol (the most readily available sample for analysis) may not be indicative of vitamin A status, and other 13 methods must be used to evaluate vitamin A status. There are other methods of assessing vitamin A status including serum RBP concentrations, plasma RBP to TTR ratios, liver vitamin A concentrations, relative dose response tests, stable isotope tracers, and early symptoms of deficiency (82). Each method of analysis has advantages and disadvantages. The role of vitamin A in vision leads to symptoms of deficiency involving the eye, such as kertinization of the eye layers, Bitot spots and xerophthalmia. Night blindness, or poor adaptation to levels of low light, is the first sign of vitamin A deficiency but is commonly underreported and undiagnosed. As vitamin A deficiency progresses, more severe eye symptoms develop that can cause permanent visual impairment. Keratinization and Bitot spots are reversible by vitamin A supplementation, but xerophthalmia is irreversible (82). Vitamin A and Innate Immunity Vitamin A helps to maintain an effective innate immune system. Vitamin A is essential for maintenance of mucosal lining and skin integrity. Vitamin A is also required for maintaining optimal innate immune cell numbers and/or lytic activity. Through transcriptional regulation, vitamin A regulates immune cell differentiation, maturation, apoptosis, and function as well as maintains effective barriers. Vitamin A-deficient individuals have impaired mucosal barriers. Mucus is an important coating for trapping and preventing pathogens from entering or damaging the delicate mucosal tissues. Vitamin A is required for the production of mucus and mucus glycoproteins by goblet cells and maintenance of goblet cell numbers (83, 84). The skin of vitamin A-deficient populations is keratinized and increased in thickness (85). The increased thickness of skin does not directly lead to increased susceptibility to infection. 14 However, increased kertinization makes the skin more fragile leading to decreased skin integrity and increased susceptibility to infections caused by abrasions. Natural killer (N K) cells are granular lymphocytes responsible for innate defenses to viral and intracellular infections. Basal NK cell numbers and activity are reduced in vitamin A-deficient animals, but are restored upon supplementation of vitamin A (86-89). Basal NK cell lytic efficiency or the ability of each NK cell to lyse a target is not impaired in vitamin A-deficient animals (90). In addition, upon stimulation with polyinosinic:polycytidylic acid there is no difference in the IFN-y production, cell proliferation, or lytic efficiency of NK cells from vitamin A-deficient and vitamin A- sufficient animals (89). In summary, in vivo NK cells are depleted in vitamin A-deficient animals, but the ability of NK cells to respond upon stimulation is unaffected by vitamin A deficiency. Macrophages are mononuclear phagocytes capable of ingesting and killing microbes within phagolyosomes through the action of lytic peptides and degradative enzymes. Macrophages also present antigens of the ingested microbes to T cells and secrete cytokines and chemokines to initiate the adaptive immune response and increase local inflammation (1). The effect of vitamin A on macrophage function is contradictory. In pathogen-free mice, vitamin A deficiency increased macrophage numbers in secondary lymphoid tissues but also impaired delayed-type hypersensitivity, or cell-mediated immune responses (91 ). Vitamin A supplementation has been reported to increase macrophage functions in vivo and in vitro (92-95). However, phagocytosis of antibody opsonized cells was impaired due to decreased expression of receptors for antibody on human macrophages cultured in vitamin A-deficient medium (96). Pro-inflammatory 15 cytokines produced by macrophages [interleukin (IL) -1 , IL-12, and tumor necrosis factor (TNF) -or)] are increased and regulatory cytokines, including IL-10, are decreased in vitamin A-deficient populations (97-101). Although the cytokines that are increased in vitamin A deficiency are normally associated with induction of oxidative pathogen killing, in the case of vitamin A deficiency, they are ineffective at stimulating the pathogen killing mechanisms. Supplementation of vitamin A increases the response to these cytokines and leads to effective pathogen killing (102—105). Therefore, vitamin A deficiency increases macrophage—mediated inflammation, decreases oxidative burst killing, but phagocytic function remains controversial. There are several pathways for triggering macrophage phagocytosis, and the effect of vitamin A on each specific pathway has not been elucidated. Vitamin A deficiency increases granulocyte numbers (106, 107). In addition, Kuwata et al. showed that vitamin A-deficient SENCAR mice had a marked significant increase in neutrophil cell numbers in the spleen, peripheral blood, and bone marrow due to impaired apoptosis (108). However, the function of granulocytes is decreased in vitamin A deficiency (109). The chemotaxic, phagocytic, and oxidative burst functions of neutrophils of vitamin A-deficient rats were significantly decreased compared to neutrophils from vitamin A-sufficient rats (1 10). Therefore, despite increased numbers, the ability of neutrophils to control early infections and induce a pro—inflammatory state is depressed in vitamin A deficiency. In summary, vitamin A deficiency impairs the innate immune system through decreasing mucosal barriers and skin integrity. Interestingly, myeloid innate immune cells (neutrophils and macrophages) are increased in vitamin A deficiency and lymphoid 16 innate immune cells (NK cells) are decreased. Vitamin A is required for normal hematopoiesis, particularly myelopoiesis, through transcriptional regulation of genes for differentiating bone marrow progenitor cells (1 1 1). Although myeloid innate immune cells are increased in vitamin A-deficient populations, the function of innate immune cells is impaired either at baseline or in response to stimuli resulting in increased susceptibility to and severity of infections. The impaired innate immune system of vitamin A-deficient populations results in uncontrolled infections and exacerbated inflammation with potential downstream effects in the adaptive immune system. Vitamin A and Adaptive Immunity Vitamin A has important functions in the adaptive immune system (1 12). In vitro assays have been used to determine direct effects of vitamin A on both B and T lymphocytes. However, the in vivo effects of vitamin A on adaptive immune responses are the combination of direct and indirect effects on lymphocyte populations. Vitamin A, as a transcription factor ligand, affects the responses of B and T lymphocytes, but the effects of vitamin A on APCs and other cells can alter the cytokines produced and thereby alter the B and T cell responses as well. Vitamin A has effects on the proliferation, differentiation, maturation, apoptosis, and other functions of adaptive immune cells. Vitamin A does not alter total T lymphocyte numbers of pathogen-free animals (91). However, specific subsets of T cells are altered in vitamin A—deficient populations. Cytotoxic T lymphocytes (CTLs) are antigen-specific cells that produce perforin and granzyme to induce apoptosis of cells infected with intracellular pathogens (1). Delayed- type hypersensitivity responses, CTL-mediated, are impaired in many, but not all vitamin 17 A—deficient populations (91, 113-115). IL-2 is an autocrine growth factor for CTLs and vitamin A upregulates IL-2 receptor mRNA and protein (116-119). The direct effect of vitamin A on CTL function has not been well established and relied primarily on experiments with Th1 cell responses. Vitamin A deficiency skews immune responses to a Th1 bias. The Th1 cytokines IF N-y and IL-12 are constitutively synthesized in vitamin A-deficient mice and supplementation with all—trans retinoic acid decreased IFN-y synthesis (97). In addition, a severe influenza infection of vitamin A-deficient mice showed decreased influenza specific IgA levels and increased influenza specific IgG levels compared to vitamin A- sufficient control animals (120). In similar experiments, influenza infections of mice supplemented with high doses of vitamin A had increased influenza specific IgA, IL-1 0, decreased serum IgG, and IFN-y (120, 121). Production of IgG augments a Th1 response through antibody-dependent cellular cytotoxicity. The depressed Th2 cytokines of vitamin A-deficient populations are the combined result of decreased Th2 cell stimulation and the cross—regulation of the increased Th1 cytokine IFN-y (122). Vitamin A-deficient populations have impaired antibody responses to T- dependent antigens (89, 91, 114, 120, 123-130). Antibody responses are produced through the cooperation of APCs, B cells, and Th2 cells. Vitamin A deficiency does not have direct effects on B cells; cell numbers, T-independent antibody responses, and antibody responses stimulated with Th2 cells from a vitamin A-sufficient mouse were unaltered in vitamin A-deficient populations (89, 1 14, 1 15, 131). Vitamin A supplementation increases the mRNA of IL-4 and IL-5, the classical Th2 cytokines, and decreases IFN—y, while vitamin A deficiency decreases IL-4 (97, 122, 132). Despite 18 unaffected antibody production per B cell, the clonal expansion of B cells is impaired in vitamin A-deficiency (123). In addition, Th2 cells are decreased in vitamin A-deficient mice and supplementing the mice with vitamin A restored Th2 cell numbers (131). Therefore, impaired antibody responses in vitamin A-deficient populations are primarily due to impaired Th2 cell help. Vitamin A is important for antibody class switching. At mucosal sites, IgA is important for innate protection, but vitamin A deficiency decreases mucosal IgA (120, 124, 126-130). In addition, B cell numbers are not altered in vitamin A-deficient populations (1 15). Therefore, the effect of vitamin A on B lymphocytes is primarily a result of impaired Th2 cell help and not directly on B cells. The depressed IgA synthesis of vitamin A-deficient populations is due to the combination of altered cytokine environment and impaired lymphocyte homing. The role of DCs in establishing the cytokine environment and directing lymphocyte homing allows for the discussion of depressed B cell function to continue in the vitamin A and DC section below. Vitamin A deficiency increases Th1 responses with unclear CTL and macrophage defects, while decreasing Th2 responses without altering intrinsic B cell function. The altered cytokine environment and cell-cell communication skews the balance of T cells in favor of Th1, leading to aberrant antibody and cell-mediated immune responses. Vitamin A does have direct effects on T lymphocyte proliferation, but T cells also require stimulation from APCs in which vitamin A may also have direct effects. Vitamin A and DCs Vitamin A, as a steroid hormone transcription factor, regulates hematopoiesis (111). Hoag and Hengesbach showed that murine bone marrow cells cultured in 19 granulocyte/macrophage-colony stimulating factor (GM-CSF) differentiated into DCs in the presence of vitamin A or RAR agonists (133). However, the bone marrow cells cultured in medium containing GM-C SF and charcoal dextran filtered serum (depleted of vitamin A) or RAR antagonists generated greater numbers of granulocytes and fewer DC 5 (133). Human bone marrow cultures also increased differentiation of myeloid progenitors from CD34+ stem cells in the presence of all-trans retinoic acid (134). Therefore, in vitro vitamin A induces differentiation from CD34+ stem cells to myeloid progenitors to myeloid DCs. Without vitamin A, hematopoietic stem cells differentiate into myeloid progenitors at a slower rate and increase production of granulocytes instead ofmyeloid DCs. In vivo, SENCAR mice consuming a vitamin A-deficient diet had dramatically increased myeloid cells in the spleen and bone marrow. The myeloid cells were primarily granulocytes, specifically polymorphonuclear neutrophils. However, the authors did not characterize the DCs of these mice (108). The effect of vitamin A on the in vitro differentiation of bone marrow progenitors combined with the observed in vivo increase in granulocytes in vitamin A deficiency leads to the hypothesis that in vivo vitamin A-deficient animals would have decreased myeloid DCs and increased neutrophils. Further, vitamin A-deficient populations have impaired Th2 antibody responses due to decreased myeloid DCs. Particularly important in intestinal tissue, DC 5 express enzymes for the metabolism of vitamin A. Retinal dehydrogenase, the enzyme responsible for converting retinal to retinol, is expressed in mucosal tissue DCs (135). Addition of vitamin A to DCs in cultures leads to the increased expression of the vitamin A metabolic enzymes in 20 DCs and adoptive transfer of these DCs leads to homing of T and B lymphocytes to the gut associated lymphoid tissue (135-138). The combined effects of impaired mucosal homing of lymphocytes and decreased cytokines, specifically IL-10, in vitamin A deficiency explain the depressed IgA levels of vitamin A-deficient populations. Therefore the role of vitamin A in DC function, particularly in DCs of the gastrointestinal tract and other mucosal sites, is critically important for intestinal immune homeostasis. Vitamin A can regulate the transcription of cytokine genes. Generally, in the presence of vitamin A, the Th1 cytokines are inhibited while the Th2 cytokines are increased. However, without vitamin A, Th1 cytokines are increased while Th2 cytokines are decreased. Thus, one may speculate that vitamin A regulates the differentiation of myeloid progenitors leading to skewed populations of APCs responsible for stimulating adaptive T lymphocyte responses. Experiment Rationale Vitamin A deficiency affects about 125 million children worldwide each year (76). Vitamin A deficiency is not as prevalent in the US. compared to other countries, but is still a major nutrient concern, especially in preschool-age children and pregnant women (77). Despite numerous supplementation programs targeting vitamin A-deficient populations, vitamin A deficiency induced morbidity and mortality remains a global concern. Dietary depletion of vitamin A in mice leads to increased neutrophils in the spleen, bone marrow, and peripheral blood, and Th1 biased immune responses (97, 108, 120, 122). Bone marrow cell cultures stimulated with GM—CSF, in medium depleted of vitamin A or in the presence of RAR antagonist, produce increased numbers of 21 granulocytes with fewer myeloid DCs, while the bone marrow cultures stimulated with GM-CSF in the presence of vitamin A produced primarily myeloid dendritic cells and few granulocytes (133). Dendritic cell (DC) subsets of vitamin A-deficient animals have yet to be characterized in vivo. DCs are the primary cell type responsible for stimulating naive T cells for adaptive immune responses. In the mouse, myeloid DCs stimulate primarily Th2 responses, while lymphoid DCs stimulate primarily Th1 responses (10, 11, 28). Plasmacytoid DCs produce type 1 interferons which aid anti—viral responses by activating NK cell killing (12, 14). All mature DC subsets have been shown to arise from a common immature DC or preDC (22). In the mouse, all DC subsets express CD1 1c on the cell surface, and MHC-II, CD8a, CD1 1b, Gr-l, and CD45RB (3220) can be used in combination to distinguish the DC subsets. We developed and employed multicolor flow cytometry to identify and quantify the DC subsets of the spleen from vitamin A-deficient (VAD) and vitamin A-sufficient (VAS) animals. We hypothesized that vitamin A deficiency would lead to significant decreases in myeloid DCs and increases in neutrophil and preDC populations, while the depletion of vitamin A would have no effect on lymphoid or plasmacytoid DC subsets. The role of vitamin A in DC phenotype and function has been thus far confined to in vitro assays. The protocol we developed has established a reliable method for analyzing DC populations of the spleen from individual C57BL/6ll mice using multicolor flow cytometry. We will provide evidence addressing the knowledge gap in the mechanism of depressed Th2 responses in vitamin A-deficient populations by quantifying 22 APCs responsible for stimulating naive T cell differentiation. Our research will provide a direct in vivo role for vitamin A in maintenance of DC populations of the spleen. The role of vitamin A in DC population homeostasis will contribute to our understanding of the mechanism for the deficiency in Th2 and T cell-dependent antibody responses observed in vitamin A-deficient populations. 23 CHAPTER 2: THE IDENTIFICATION AND ENUMERATION OF DENDRITIC CELL POPULATIONS FROM INDIVIDUAL MOUSE SPLEEN AND PEYER’S PATCHES USING FLOW CYTOMETRIC ANALYSIS Duriancik DM and Hoag KA. 2009. Cytometry, part A. 75:951-9. Supplemental Figures and Tables published online as supporting material for this manuscript can be found in Appendix I. 24 ABSTRACT Dendritic cell (DC) research currently involves pooling of tissues from multiple animals followed by enrichment techniques to obtain sufficient numbers of DCs for analysis. Enrichment techniques take advantage of DC adherence, buoyant density properties, and/or positive or negative selection of cell populations using monoclonal antibodies. However, enrichment techniques may significantly change the maturation and/or activation status of DCs or selectively eliminate one or more subpopulations of DCs. To overcome these drawbacks, we designed a multicolor flow cytometric technique for simultaneous analysis of DC populations from tissues of individual mice. The spleens and Peyer’s patches were mechanically and enzymatically digested, then incubated with a panel of 6 monoclonal antibody-fluorochrome direct conjugate reagents. A BD® Biosciences LSR II flow cytometer and FCS Express® software were used to identify 3 subtypes of mature DC 3 (myeloid, lymphoid, and plasmacytoid), precursor DCs, polymorphonuclear neutrophils, B lymphocytes, and Gr-1+/CD80t+ memory T lymphocytes in the spleen. Likewise, we also identified these DC subpopulations and B lymphocytes in the Peyer’s patches. The three key parameters in analysis of the DC populations were bi-exponential plotting in data analysis, collection of a minimum of 50,000 total events, and accurate color compensation. This procedure to analyze DCs from individual mice can lead to further understanding of the role of DCs in many other model systems as well as better understanding of how dietary or physiological factors may affect in vivo DC homeostasis. 25 INTRODUCTION Dendritic cells (DCs), first described by Steinman and Cohn in 1973, are antigen presenting cells sparsely distributed throughout the body (2). The lineage development of DCs remains vague due to the plasticity of DCs. In the mouse, myeloid DCs primarily stimulate antibody-mediated T helper 2 responses and lymphoid DCs primarily stimulate cell-mediated T helper 1 responses (11, 139). Plasmacytoid DCs secrete large amounts of type 1 interferons to augment anti-viral responses (12-14). The mature DCs (myeloid, lymphoid, and plasmacytoid) have been shown to share a common DC precursor (22). However, others have reported two precursor populations, one for plasmacytoid DCs and another for both myeloid and lymphoid DCs (140, 141). At mucosal sites, yet another DC subpopulation expressing CD103 is responsible for stimulating T regulatory cell maturation (142, 143). The lifespan among the various subpopulations varies from about 1.5 days to 3.0 days for lymphoid DCs and myeloid DCs to about 9 days for plasmacytoid DCs (8, 19). Therefore, hematopoietic precursors in the bone marrow produce new DCs to replace the dying DCs. Typically, the spleen has been reported to possess less than 1% total DCs, or 1-3 x 105 DC per spleen, with a ratio of 3 to 1 of myeloid to lymphoid DCs (3, 8). However, the authors’ used the DC properties of low buoyant density and adherence to glass to isolate and enrich for DCs (3). A variety of factors may influence the number of DC 3 in a tissue or animal at a given time, including infection, chronic disease states such as diabetes, hormones such as estradiol, nutrients such as vitamins A and D, as well as others (133, 144-148). However, previous methods used to analyze DCs have used enrichment techniques from pooled samples, either multiple tissue or animal sources, to 26 obtain sufficient numbers of cells for analysis leading to potentially high variability and inaccurate DC number assessments. Despite numerous publications quantifying DCs in tissues and animals, accurate analysis relies on in vivo expansion of DC prior to isolation, enrichment techniques, or positive or negative selection procedures (145, 149-151). Also DC identification using flow cytometry has been documented in Cytometry, Part A (152-155). However, these publications have either identified only one of the multiple DC subpopulations or focused on a specific tissue such as the lung, blood or bone marrow, not all identifiable DC populations present in the spleen or Peyer’s patches. Due to the variability of immune cell populations among animals and the high cell loss associated with various enrichment techniques, we employed 6 monoclonal antibody- (mAb) fluorochrome direct conjugates to identify all resident DC subpopulations of the spleen and Peyer’s patches of individual, healthy mice. The use of multiple fluorochromes to detect low percent cell populations requires digital compensation with fluorochrome-labeled compensation beads, bi- exponential scaling on analysis plots, and the collection of minimally 50,000 flow cytometric events. Here we describe the gating strategy used to detect DC populations, as well as the percent and total cell numbers observed in individual, healthy, male and female C57BL/6J mice. METHODS Animals Male and female mice, C 57BL/6J, were obtained from Jackson Laboratories (Bar Harbor, ME, USA) and used as breeder pairs. Pups were weaned at 3 weeks of age and fed ad libitum commercial solid pellets (Harlan Teklad 22/5 Rodent Diet #8640). At 12 27 weeks of age, the mice were killed by C02 asphyxiation. All procedures were in accordance with Michigan State University Laboratory Animal Resource guidelines, and animals were housed in a facility accredited by the Association for Assessment and Accreditation of Laboratory Animal Care. Tissue Isolation The spleen and Peyer’s patches were excised and placed in ice cold calcium and magnesium free Hank’s balanced salts solution (HBSS, Gibco, Carlsbad, CA, USA). The tissues were mechanically minced with scissors and enzymatically digested with Spleen Dissociation Medium (Stem Cell Technologies, Vancouver, BC, Canada) following manufacturer’s instructions. The tissues were filtered through 70pm mesh (Sefar America Inc, Depew, NY, USA). Cell suspensions were centrifuged at 1200 RPM, 4°C, 10 minutes to pellet. All subsequent centrifugations were at 1200 RPM, 4°C, 10 minutes. The spleen cells were resuspended and incubated for 2 minutes in PharmLyse (BD Biosciences, San Jose, CA, USA) on ice. The spleen cells were diluted with HBSS and centrifuged to pellet. The spleen and Peyer’s patch cells were resuspended in 5mL fluorescence activated cell-sorting (F ACS) buffer [0.1% sodium azide (Fisher Scientific, Pittsburgh, PA, USA), 1.0% fetal bovine serum (Hyclone, Logan, UT, USA) in Dulbecco’s phosphate buffered saline, pH 7.4-7.6, sterile filtered]. The cells were counted on a hemacytometer using trypan blue exclusion dye (Biowhittaker, Walkersville, MD, USA). One million cells were dispensed into each tube for subsequent antibody-fluorochrome staining and analysis. 28 Antibody Staining The cells were centrifuged to pellet and incubated with 5ug of anti-FcRyII/III antibody (2.4G2 hybridoma) on ice for 10 minutes. A master mix of all 6 mAb- fluorochrome conjugates (Table 1; BD Biosciences, San Jose, CA, USA) was prepared with lug of each mAb-fluorochrome conjugate per one million cells in SOuL total volume. The cells were incubated with the master mix at 4°C for 30-60 minutes in the dark. Cell suspensions were washed with lmL of F ACS buffer, centrifuged to pellet, resuspended in 0.5mL FACS buffer, and transported to the MSU Flow Cytometry Core Facility on ice and in the dark. The cells were run the same day and were only on ice for 1-2 hours prior to flow cytometer runs. Flow Cytometry Acquisition & Analysis An LSR 11 (BD Biosciences, San Jose, CA, USA) flow cytometer (Supplemental Figure l) was used to collect at least 50,000 total events. The LSR II instrument parameters and general compensation matrix are shown in Supplemental Tables 1 and 2, respectively. Compensation was preformed prior to each data collection using BD Bioscience anti-rat K CompBeads according to manufacturer’s recommendations. A representative figure of the fluorochromes requiring the most compensation is shown in supplemental figure 2. Anti-CDl4-PE was substituted for anti-CD1 lc-PE because the CompBeads do not bind a hamster antibody. We assumed an equal fluorochrome to protein ratio for both anti-CD14-PE and anti-CD1 lc-PE. The events were collected and post-acquisition analysis was performed using FCS Express version 3 (De Novo Software, Los Angeles, CA, USA). 29 Flow cytometric analysis gating strategies Table 2 summarizes the cell surface antigens that were utilized to identify the immune cell populations. The gating strategies for spleen and Peyer’s patch cells are shown in Figs. 1 and 2, respectively. The forward scatter and side scatter plot is used to distinguish the live cells from dead cells, debris, and doublet cells (Figs. 1A & 2A). For spleen cell populations, the Gr-l-PeGC-Cy5.5 and CD8or-PE-Cy7 plot is used to distinguish the memory Gr-l+/CD8(1+ T cells and granulocytes, since granulocytes do not express CD80t (Fig. 18). Memory CD8+ T lymphocytes have been shown to express Ly-6C, which along with Ly-6G is recognized by the anti-Gr-l antibody (156, 157). Granulocytes (Gr-1+/CD80t' gate in Fig. 1B) are confirmed as polymorphonuclear neutrophils (PMN) by the expression of CD1 1b in the histogram of CD11b-APC-Cy7 (Fig. 1C). The histogram of CD1 lc-PE (Fig. 1D) is used to identify all DC populations, and the histogram of MHC-II-FITC (Fig. IE) is used to distinguish preDCs and mature DC 3, with preDCs lacking MHC-II expression and mature DCs exhibiting MHC-II expression. In the mouse, all DC populations express CD1 1c (8, 11). Mature mouse DC 5 (CD1 lcVMHC-IIJr gate) are divided into three distinct subpopulations by the expression of Gr-l and B220 (plasmacytoid DCs; Fig. 1F), CD80. (lymphoid DCs) and CD1 1b (myeloid DCs; Fig. 10) (8, 11). The preDCs (CD1 1c+/MHC-II' gate) are confirmed by the expression of B220 and CD1 1b in the plot of BZZO-APC and CD1 lb-APC—Cy7 (Fig. 1H; (22)). The histogram of Gr-l-PeGC- Cy5.5 is used to identify potential B cells, which are negative for the expression of Gr-l (Fig. 11). Any residual cells expressing Gr-l-PeGC-Cy5.5 not gated as PMN or plasmacytoid DC will be eliminated (Fig 11). The B cells are further confirmed by the 30 expression of BZZO-APC and variable expression MHC-II-FITC (Fig. 1.1). The expression of MHC-II on B cells is indicative of B cell antigen presentation. As cell populations are identified, exclusion gates using “not” Boolean logic are used to eliminate events from subsequent plots (Fig. 1K and supplemental figure 3). For example, granulocytes and Gr-l + memory T cells are identified and excluded using the minus granulocyte or Gr-l T gate with the Boolean logic not (granulocyte or Gr-l T). The mAb-fluorochrome combination we utilized is limited to the identification of the cell populations described above. Although the spleen contains other cell populations, our gates do not identify all T lymphocytes, natural killer cells or macrophages, but they are likely not contained in our final DC populations. A flow diagram of the gating strategy is shown in supplemental figure 3 for clarity of Boolean logic employed. The gating strategy of the Peyer’s patch cells is shown in Fig. 2. Since Peyer’s patches do not have the blood filtering function of the spleen, PMNs should not be present in appreciable numbers and thus are excluded from the gating strategy. Therefore, the plot of Gr-l-PeGC-Cy5.5 and CD80t-PE-Cy7 used to identify granulocytes and memory T cells in the spleen is not used in the Peyer’s patch cell analysis. The histogram of CD1 lc-PE is used to identify potential DCs (CD1 1c+; Fig. 2B), and the histogram of MHC-II-FITC (Fig. 2C) is subsequently used to distinguish preDCs (MHC-II') and mature DCs (MHC-IF). The mature DCs (CD11c+/MHC-II+) are divided into the 3 distinct subpopulations by the expression of Gr-l and 8220 (plasmacytoid DCs; Fig. 2D), CD8ot (lymphoid DC) and CD1 1b (myeloid DCs; Fig. 2E). A fourth mature DC population not expressing the markers employed in the analysis is identified (“Other DC”; Fig. 2E). The preDCs (CD1 lcVMHC-II' gate) are confirmed by 31 the expression of 8220 and CD1 1b in the plot of BZZO-APC and CD1 lb-APC-Cy7 (Fig. 2F). The histogram of Gr-l-PeGC-Cy5.5 is used to identify potential B cells (Fig. 2G). The B cells are further confirmed by the expression of B220 and variable expression of MHC-Il (Fig. 2H). B cells do not express Gr-l, but do express B220. The expression of MHC-II on B cells is indicative of B cell antigen presentation. As cell populations are identified, exclusion gates using “not” Boolean logic are used to eliminate events from subsequent plots (Fig. 21). There are other immune cell populations present in Peyer’s patches (T lymphocytes), but our mAb-fluorochrome cocktail only allows for the identification of the cell populations described above. Population calculations and statistical analysis The total cell numbers for each population per tissue were obtained by multiplying the percentage of the respective gate from the flow cytometry analysis by the total cells (for spleens) or total cells per Peyer’s patch counted on a hemacytometer. Prism (GraphPad software , San Diego, CA) was used to perform a student’s t-test to detect statistical significance (p< 0.05) between male and female animals for each cell population percentage and total cell numbers. RESUL TS Animal characteristics At 12 weeks of age, the male and female mice had an average body weight of 25g and 19g, and spleens weights of 0.06g and 0.10g, respectively (Table 3). One female mouse had an unusually large spleen weighing 0.24g. Eliminating this outlier caused the average spleen weight for female mice to decrease to 0.07g. However, the female mouse 32 with an abnormal spleen weight contained similar number of nucleated cells, suggesting the excessive weight may have been due to red blood cell retention in the spleen. The nucleated cell number per spleen was 1.19 x 108 and 1.36 x 108 for males and females, respectively (Table 3). The average number of Peyer’s patches excised was 7 for both males and females, with a minimum of 5 and a maximum of 9. The number of cells per Peyer’s patch was 8.40 x 105 and 8.19 x 105 for males and females, respectively (Table 3). Average immune cell numbers obtained from healthy C57BL/6J mouse spleen and Peyer’s patches We used the techniques described above to determine the percent and total DC subpopulation numbers (Table 4) and other non-DC immune cell populations (Table 5) present in the spleens and Peyer’s patches of healthy male and female C57BL/6J mice. Due to the varying numbers of Peyer’s patches excised per animal, the Peyer’s patch data is shown as cells per Peyer’s patch and not total cell numbers. The total mature and preDCs represent about 4% of the spleen and 3.75% of the Peyer’s patches in both male and female mice. Myeloid DCs are the predominant DC subpopulation present in spleen, representing about 2% of the total spleen cells in both male and female mice (Table 4). In the Peyer’s patch, myeloid DCs compose only about 1% of the total cells in both males and females. The lymphoid DCs are about 0.7% of the spleen cells in both male and female mice, and about 0.3% and 0.4% of male and female Peyer’s patch cells, respectively. Female mice have a significantly higher percentage of lymphoid DCs in the Peyer’s patches than male mice (p=0.035). Plasmacytoid DCs are only about 0.2% of the total spleen cells in both males and females, but predominate in 33 the Peyer’s patches, representing about 1.3% of male and 1.1% of female total Peyer’s patch cells. The preDCs represent approximately 1% of total cells in both spleen and Peyer’s patches of both male and female mice. “Other” DCs are not present in the spleen, but represent about 0.2% of the Peyer’s patch cells. On average, there are 2.60 x 106 myeloid DCs, 8.02 x 105 lymphoid DCs, 2.38 x 105 plasmacytoid DC 3, and 1.51 x 106 preDCs in male mouse spleens (Table 4). Female mouse spleens contained an average of 2.83 x 106 myeloid DCs, 8.89 x 105 lymphoid DCs, 2.62 x 105 plasmacytoid DCs, and 1.47 x 106 preDCs. In each Peyer's patch of the males, there are an average of 8.83 x 103 myeloid DCs, 2.25 x 103 lymphoid DCs, 9.98 x 103 plasmacytoid DCs, 8.07 x 103 preDCs, and 1.89 x 103 “other” DCs. In each female mouse Peyer’s patch, there are an average of 9.50 x 103 myeloid DCs, 3.20 x 103 lymphoid DCs, 8.75 x 103 plasmacytoid DCs, 8.17 x 103 preDCs, and 1.82 x 103 “other” DC 5. B cells are the majority population in both the spleen (66% in males, 62% in females) and Peyer’s patches (82% in males, 78% in females; Table 5). In the Peyer’s patches, male mice have a significantly higher percentage of B cells than female mice (p=0.008). The PMNs represent only about 2% of the spleen cells in both the male and female mice, but were not quantified in the Peyer’s patches. Likewise, the Gr—1+/CD80t+ memory T cells were not quantified in the Peyer’s patches, but represent about 3% of the cells in the spleen (Table 5). In the spleen, male mice have a significantly higher percentage of Gr-1+/CD80t+ memory T cells than female mice (p=0.042). The spleens of male and female mice contain averages of 2.76 x 106 and 3.00 x 10° PMNs, respectively (Table 5). Memory Gr-1+/CD80t+ T cells average 3.71 x 106 and 34 3.61 x 106 in the spleens of male and female mice, respectively. The male and female mouse spleens contained 7.91 x 107 and 8.38 x 107 B lymphocytes, respectively, while there are 6.87 x 105 and 6.43 x 105 B lymphocytes per Peyer’s patch in male and female mice, respectively. DISCUSSION Using multiple mAb-fluorochrome conjugates in flow cytometry, we were able to identify seven immune cell populations of the spleen and five immune cell populations of the Peyer’s patches. Of note, all four mouse DC spleen subpopulations could be quantified, and our analysis suggests a fifth relevant mature DC population is present in the Peyer’s patches. The cell populations of the spleen included myeloid DCs, lymphoid DCs, plasmacytoid DCs, preDCs, PMNs, B cells, and Gr-l+/CD80t+ memory T cells. There have been reports of both one common or two separate precursor DC populations present in the mouse (22, 140). Our staining and gating strategy can identify either of these preDC populations, but here we chose to identify one common precursor DC population as described by del Hoyo et al. (2002). In the Peyer’s patches, the preDCs, myeloid DCs, lymphoid DCs, plasmacytoid DCs, and B cells were identified with the mAb-fluorochrome conjugates and gating strategy used. The mature DCs present in the Peyer’s patches that do not stain for CD80t or CD1 1b and which we classify as “other” DC could be CD103+ DCs, but we have not verified this (Figure 2E; (158)). The expression of CD80t and CD1 1b on CD103+ DCs is controversial, as lung and bronchial lymph node CD103+ DCs have been reported to express low levels of CD1 lb (159, 160) but not CD80L (159), while spleen marginal zone CD103+ DC have been reported to express CD80t (161). Thus, further experimentation would be needed to identify the 35 “other” DC subpopulation composing approximately 6% of the Peyer’s patch DCs. We suggest that DC subset analysis protocols designed for mucosal immune tissues should contain antibodies to CD103 as well as CD1 1b and CD80L. This could be added to our existing protocol since detection channels remained available on the LSR II flow cytometer. The protocol employed is novel in that single tissues from individual animals were used for analysis instead of pooling multiple cell sources or use of any enrichment techniques. In the past, enrichment techniques have led to the accidental depletion of DC subsets, in particular plasmacytoid DCs. B220, or CD45RB, was used to negatively select B cells leading to the simultaneous depletion of plasmacytoid DCs (16), yet this negative selection technique is still marketed for spleen DC enrichment [BD Biosciences IMagTM cell separation system technical document (http://www.bdbiosciences.com/pdfs/brochures/04-7900030-2-Al .pdf; accessed 05/21/2009)]. Therefore, the complex expression pattern of DC subsets limits the value of negative selection protocols based on a combination of surface proteins. The low percent of DCs, particularly in the steady-state, has led to pooling of multiple tissues or single tissues from multiple animals to obtain sufficient cell numbers for enrichment and/or analysis. However, DCs from various tissues are heterogeneous and variable depending on the tissues pooled. Lymph node analysis performed on lymph nodes pooled from various areas of the body would likely be misleading. CD103+ DCs are restricted to mucosal sites, thus pooling of mucosal lymph nodes with other lymph nodes would be inadvisable. In addition, many factors can affect individual mouse 36 immune systems, including genetics, glucocorticoids (stress), diet, and others. Therefore, inter-animal variability can be high despite numerous controls and use of inbred strains. Many variables can affect DC percentages and total cell numbers in a tissue. Infections cause DCs to mature, home to secondary lymphoid tissues, and proliferate. However, the maturation, homing, and proliferation of DCs are site specific to the secondary lymphoid organ of that area of the body. A respiratory infection will not cause systemic changes in DCs, but the respiratory draining lymph nodes will have vastly greater numbers of mature DCs. Due to the effect of estrogen on DCs, there may be differences in DC percentages and total cell numbers between sexes of animals (162, 163), but our data show few gender differences. In addition, animal age has been shown to modify DC populations (164, 165). Finally, the strain of the animal may also affect the percent and total DC numbers (166). BALB/cJ and C57BL/6J animals appear to have similar DC percentages and total cell numbers, but F VB/NJ animals of the same sex and age have significantly fewer spleen DCs and reduced capacity of bone marrow to generate DC in vitro (Duriancik, Lackey, and Hoag, unpublished data). Thus, we conclude that it is extremely important to confine DC subpopulation analysis to single tissues from individual animals, without inclusion of enrichment techniques prior to flow cytometric analysis. The strengths of the protocol presented include the analysis of DCs from single tissues of individual animals, the lack of any cell enrichment techniques, the identification and analysis of DC subpopulations using a single cocktail of mAb— fluorochrome conjugates, and the use of direct mAb—fluorochrome conjugates. The use of direct mAb-fluorochrome conjugates decreases the incubation time because there is 37 only one incubation step with the antibodies. The use of indirect conjugates may increase the sensitivity of the fluorochromes, but the increased time for multiple incubations and washing steps increases the chance for cell loss through death or DC activation due to contamination. A single cocktail of mAb-fluorochrome conjugates to identify all the DCs allows the identification of all DC subpopulations in a single tube decreasing staining variability due to different cells per tube or mAb-fluorochrome conjugates per tube. Also using a single tube of cells and elimination gates decreases the possibility of counting cells as multiple populations due to expression of multiple antigens. The lack of any cell enrichment procedure decreases cell loss, the potential of DC activation, and unintentional selection or deletion of particular cell populations. The variability of DC populations associated with different tissues including the spleen, Peyer’s patches and other mucosal sites, various draining lymph nodes, and other DC sources is eliminated by analyzing single tissues. Analysis of individual animals also increases statistical power by allowing for analysis of inter-animal variability within treatment groups. The protocol described also has minor limitations including potential cell loss from multiple centrifugations, digital flow cytometric setup and analysis, sophisticated post-acquisition gating analysis and utilization of exclusion gating, and the collection of at least 50,000 events. Increased centrifugations may lead to increasing cell losses, especially because of the low buoyant density of DCs. Digital electronics of the flow cytometer are required to accurately establish the compensation matrix, and bi- exponential plotting is required due to the compensation of the fluorochromes forcing some events to fall below zero (167, 168). Bi-exponential plotting allows for better separation of populations because data falling below zero is not condensed on either axis 38 (168). Better separation of populations leads to more accurate gating and identification of populations. The complex compensation and low cell numbers expressing some of the mAb—fluorochromes, such as CD1 1c, requires the use of compensation beads to establish the compensation matrix. Without the compensation beads too many events would be required to accurately detect the positive and negative peaks of some of the fluorochromes leaving few cells left for analysis. Due to the complex expression profile of DC subpopulations, a complex gating strategy must be employed to effectively identify all of the individual DC populations. Not only must bi-exponential plotting be used to create an appropriate cell population separation, but exclusion gates should also be used to eliminate the potential of other cell populations from contaminating true populations. If exclusion gates are not used, then the potential for other events expressing a similar profile of markers could skew the population numbers. The collection of at least 50,000 events is required because of the low percent of DCs in any tissue. If the spleen contains less than 5% of cells as total DCs, collection of 50,000 events should show about 2500 total DCs, whereas collection of only 10,000 events should show only 500 total DC 3. The separation of 2500 DCs compared to 500 DCs into 4 subpopulations allows for easier identification and analysis of DC subpopulations. However, the collection of 50,000 events increases the amount of time the sample must be run through the flow cytometer and requires a higher cell number to start. In summary, our protocol for identification and enumeration of DC subpopulations from the spleen and Peyer’s patches of individual uninfected C57BL/6J mice is an efficient method for analyzing DC populations. Several advancements in multicolor flow cytometry techniques were used in development of our protocol and 39 gating strategies (167-170). The lack of enrichment techniques decreases the associated potential cell loss and variability. The accurate identification and enumeration of DCs provides a technique that will allow for better analysis of DCs in steady-state and inflammatory conditions. Finally, we present a protocol that could be consistently adapted by multiple investigators, allowing for comparison of results from many studies performed in different laboratories. A CKNO WL EDGEMEN rs We thank the MSU Flow Cytometry Research Technology Support Facility director, Dr. Louis King for his assistance in developing the research protocol. 40 Figure 2.1. Gating strategy to identify dendritic cells and other immune cell populations in mouse spleen. 1A, forward scatter (F SC) and side scatter (SSC) to identify live cells, live cell gate, based on cell size and granularity. 1B, contour plot of CD80t-PE-Cy7 and Gr—l-PeGC-Cy5.5 used to identify granulocytes, preGranulocyte gate, as Gr-1+/CD80t' and memory T cells, Gr-l T cell gate, as Gr-1+/CD80t+. 1C, histogram of CD1 lb-APC- Cy7 used to confirm preGranulocyte gated cells as PMN, Gr-l+/CD80t'/CD1 1b+. 1D, histogram of CD1 lc-PE used to identify DCs, CD1 lc+. 1E, histogram of MHC-II-FITC to separate DCs as preDCs, CD1 1c+/MHC-II', and mature DCs, CD11c+/MHC-II+. 1F, contour plot of BZZO-APC andiGr-l-PeGC-Cy5.5 to identify plasmacytoid DCs, CD1 lc+/MHC-II+/B220+/Gr-l +. 1G, contour plot of CD8a-PE-Cy7 and CD1 1b-APC- Cy7 to identify non-plasmacytoid DCs as lymphoid DCs, CD1lc+/MHC-II+/B220'/Gr-l' /CD8(1+/CDI lb', and myeloid DCs, CD11c+/MHC-II+/B220'/Gr-l’/CD8or'/CD11b+. 1H, contour plot of CD11b-APC-Cy7 and B220-APC to confirm preDCs, CD11c+/MHC-II', as CD1 lbva’iab'c/B220+. ll, histogram of Gr-l-PeGC-Cy5.5 to identify potential B cells as Gr-l', eliminating cells expressing Gr-l-PeGC-Cy5.5 not gated as a PMN or plasmacytoid DC. 1], dot plot of BZZO-APC and MHC-II-FITC to confirm potential B cells, Gr—l', as B220+/MHC-Ilva"ab'e. 1K, Boolean logic used for hierarchical gating, including exclusion gates such as minus granulocytes or Gr—l T. Numbers in parentheses indicate cell population percentage that falls within the gate. Representative data from one animal is shown. 41 <-m.m>o-aoan_ to 203353;. $20.92. e 3 Bo V'OdV OZZG iunoo <.O._._u_ =-0_.=2 <.m.m>o-eocen_ to «330.35.: lLl. 02a m:mo h 7.0 tunoo v-Mo-ad 290:) ID «run. or F00 260 9,: tunoo V‘OSJ 42 Figure 2.1 Continued. Gating strategy to identify dendritic cells and other immune cell populations in mouse spleen. 1A, forward scatter (FSC) and side scatter (SSC) to identify live cells, live cell gate, based on cell size and granularity. lB, contour plot of CD80t-PE-Cy7 and Gr-l -PeGC-Cy5.5 used to identify granulocytes, preGranulocyte gate, as Gr-l+/CD80t' and memory T cells, Gr-l T cell gate, as Gr-l+/CD80t+. 1C, histogram of CD1 1b-APC-Cy7 used to confirm preGranulocyte gated cells as PMN, Gr- 1+/CD80t'/CD1 lbl. 1D, histogram of CD1 lc-PE used to identify DCs, CD1 lc+. 1E, histogram of MHC-II-FITC to separate DCs as preDCs, CD1 lcVMHC-II', and mature DCs, CD1 lc+/MHC-II+. 1F, contour plot of BZ20-APC and Gr-l-PeGC-Cy5.5 to identify plasmacytoid DCs, CD11c+/MHC-II+/B220+/Gr-l+. 1G, contour plot of CD80L- PE-Cy7 and CD1 lb-APC-Cy7 to identify non-plasmacytoid DCs as lymphoid DCs, CD1lc+/MHC-ll+/8220'/Gr-1'/CD80t+/CD1lb', and myeloid DCs, CD1 lc+/MHC- Il+/BZ20'/Gr-l'/CD80t'/CD1 1b+. 1H, contour plot of CD1 lb-APC-Cy7 and B220-AFC to confirm preDCs, C D1 lcVMHC—II', as CD1 lbvarlab'e/8220+. ll, histogram of Gr-l-PeGC- Cy5.5 to identify potential B cells as Gr-l', eliminating cells expressing Gr-l-PeGC- Cy5.5 not gated as a PMN or plasmacytoid DC. 11, dot plot of B220-AFC and MHC-II- FITC to confirm potential B cells, Gr-l', as B220+/MHC-Ilva"ab'“. 1K, Boolean logic used for hierarchical gating, including exclusion gates such as minus granulocytes or Gr—l T. Numbers in parentheses indicate cell population percentage that falls within the gate. Representative data from one animal is shown. 43 CD8a PE—Cy7 (— I Lymphoid preDCZ fi‘ N . > 9 . >- ' e. j 0 E .l ‘ ' , ~ a a Myeloid / ‘13 0 y , CD11bAPC-Cy7—A 8 3220 AFC-A Gr1F’eGC-Cy55-A Bcells K g I Live Cells (73.14%) I Gr-1 T cells (2.42%) 4, E] preGranulocytes I Granulocytes (8.69%) 8220 APC-A : I Minus granulocytes and Gr-1 T MHC-Il FlTC-A : I CD110 pos (14.32%) : I Mature DCs I Plasmacytoid (0.68%) E I Minus Plasmacytoid I Myeloid DCs (5.59%) I Lymphoid DCs (4.31%) E I preDCs . I preDCZ (1.66 %) l—J I Minus all DCs E E preB cells I Bcells (55.55%) Figure 2.2. Gating strategy to identify dendritic cells and other immune cell populations in mouse Peyer’s patches. 2A, forward scatter (F SC) and side scatter (SSC) to identify live cells, live cell gate, based on cell size and granularity. 28, histogram of CD1 lc-PE used to identify DC 3, CD1 1c+. 2C, histogram of MHC-II-FITC to separate DCs as preDCs, CD1 1c+/MHC-Il', and mature DCs, CD11c+/MHC-II+. 2D, contour plot of BZ20-APC and Gr-l -PeGC-Cy5.5 to identify plasmacytoid DCs, CD11c+/MHC- III/B220+/Gr-l+. 2E, contour plot of CD80r-PE-Cy7 and CD1 1b-APC-Cy7 to identify non-plasmacytoid DCs as lymphoid DCs, CD1 1c+/MHC-II+/B220'/Gr-l'/CD80L+/CD1lb', myeloid DCs, CD11c+/MHC-II+/BZ20'/Gr-l'/CD8or'/CD11b+, and “Other” DCs, CD11c+fMHC-Il+/B220'/Gr-l"/CD80t'/CD1lb". 2F, contour plot of CD1 lb-APC-Cy7 and BZZO-APC to confirm preDCs, CD11c+/MHC-II', as CD1 lb”“"“"'°/13220*. 20, histogram of Gr-l-PeGC-Cy5.5 to identify potential B cells as Gr-l'. 2H, dot plot of BZZO-APC and MHC-Il—FITC to confirm potential B cells, Gr-l', as B220+/1\/1HC-Ilva"ab"’. 21, Boolean logic used for hierarchal gating, including exclusion gates. Numbers in parentheses indicate cell population percentage that falls within the gate. Representative data from one animal is shown. 45 <-0n_< ONNm Noemi . _ Lira _ <.O._._n_ __-OI_2 3.3 _ ._. r c. i v-Mo-oav qr LOO iunoo <-§o-oo< o F Bo use»: con 350 (run. or FOO .lr. -it- 2 Lu VIOEd 900 iunog $099818 to V'OdV 0228 D V'OSd o__oo 9: 46 Figure 2.2 Continued. Gating strategy to identify dendritic cells and other immune cell populations in mouse Peyer’s patches. 2A, forward scatter (FSC) and side scatter (SSC) to identify live cells, live cell gate, based on cell size and granularity. ZB, histogram of CD1 lc-PE used to identify DCs, CD1 1c+. 2C, histogram of MHC-II-FITC to separate DCs as preDCs, CD11c+/MHC-II', and mature DCs, CD1 lc+/MHC-II+. 2D, contour plot of B220-APC and Gr-l-PeGC~Cy5.5 to identify plasmacytoid DCs, CD11c+/MHC- II+/8220+/Gr-l+. 2E, contour plot of CD8or-PE-Cy7 and CD1 lb-APC-Cy7 to identify non-plasmacytoid DCs as lymphoid DC 5, CD11c+/MHC-II+/B220'/Gr-l'/CD8or+/CD1 lb', myeloid DCs, CD1lc'i/MHC-Il’L/BZZOVGr-1'/CD8or'/CD11b+, and “Other” DCs, CD1lCVMHC-IIVBZZOVGFI7CD8Q7CDI1b-. 2F, contour plot of CD1 1b-APC-Cy7 and BZZO-APC to confirm preDCs, CD11c+/MHC—II', as CD1 le'a'iab'C/Bz20fi 20, histogram of Gr-l-PeGC-Cy5.5 to identify potential B cells as Gr-l'. 2H, dot plot of B220-APC and MHC-II-FITC to confirm potential B cells, Gr-l', as BZZO+/MHC-Ilva'lab'c. 21, Boolean logic used for hierarchal gating, including exclusion gates. Numbers in parentheses indicate cell population percentage that falls within the gate. Representative data from one animal is shown. 47 Count 8220 APC-A Gr1 PeGC-Cy5.5—A B cells MHC-Il FITC-A t-‘ L .l Live Cells (66.47%) E} I CD11c pos (2.58%) g I Mature DCs % . I Plasmacytoid (0.52%) I [3 I Minus Plasmacytoid 3 I Other DCs (0.68%) _ I Lymphoid DCs (0.20%) , : I Myeloid DCs (0.50%) E] I preDCs f I preDC2 (0.48%) E] I Minus all DOS [3. D preB cells I Bcells (75.71%) Table 2.1. Antibody-fluorochrome conjugate reagents employed in labeling cell suspensions for flow cytometric analysis. The fluorochromes were selected for maximum sensitivity (17] ). Antigen Clone Isotype Fluorochrome MHC-ll (l-A/l-E) 2G9 Rat Inga. K FITC CD110 HL3 Arm. Hamster IgG]. 79 PE CD80L 53-6.7 Rat Inga, K PE-Cy7 CD1 lb M1/70 Rat Ingb. K APC-Cy7 CD45RB (8220) RA3-6BZ Rat 1862a, K APC Gr-l RB6-8C5 Rat Ingb. K PeGC-Cy5.5 49 Table 2.2. Antigen expression of immune cell populations in mouse spleen and Peyer’s patches used in gating strategy. Cell Population Dendritic cells Antigen Myeloid Lymphoid Plasmacytoid PreDC CD1 lc posb P03 P03 P05 MHC-II P03 P05 P03 Negd CD1 1b Pos Neg N/A Pos/Neg CD80: Neg Pos N/A N/A Gr-l Neg Neg Pos N/A 8220 Neg Neg P03 P03 50 Table 2.2 mdPQCl a,” Anhgcn F...—————— (l)llc lllltlll ._—____ (‘Dllb- hillldf— hEhtl—_m _EEEU.2 bPosind L\;\ind Slrdlcux, L1,, \Cglnd Table 2.2 Continued. Antigen expression of immune cell populations in mouse spleen and Peyer’s patches used in gating strategy. Cell Population PMN“ B Cells Gr-VCDSa * T Cells Antigen CD1 1c N/p‘C N/A N/A MHC-ll N/A Pos/Neg N/A C D1 1b Pos N/A N/A CD80L Neg N/A Pos Gr-l Pos Neg Pos 8220 N/A Pos N/A aPMN is polymorphonuclear neutrophil. bPos indicates that the cell population is positive for the antigen. cN/A indicates that the antigen is not used to distinguish the cell population in our gating strategy. dNeg indicates that the cell population is negative for the antigen. 51 Table 2.3. Physiological characteristics of C57BL/6J mice used in the studiesa. Body Spleen Cells/Spleen Number of Cells/PPC Weight Weight PPb (g) (a) Male 24.77 3: 0.06 : 119 x 108 i 6.9 i 1.4 340 x 105 i (“27) 1'06 0'01 2.94 x 107 3.33 x 105 Female 19.03: 0.10: 1.36x108: 6.5:].1 319x105: ("26) 0'58 0'07 1.77 x 107 2.27 x 105 aAll data presented as mean i 1 SD. bPP is Peyer’s patches. CTotal cells counted for all Peyer’s patches from one animal divided by the total number of Peyer‘s patches harvested from the animal. 52 .COO—Qm Us? a: COHUDHDU HOG 0.53 mUQ MUSwO HMS“ mowmo_flcm <\Z.o duo—mm 2: E coma—smog some CE 230 .39 no 223308 3%:de -\+ owwco>~ 2 0.6« 2 1: «5 0.5 4: a. F. 0.4« 3 . 0 3 [3 p value ' Liver RAE —0.68 0.00 0.2, Age 0.24 0.00 Gender -0.39 0.00 0.1 R2 = 0.52 0.00 0.0 r 51 Gt 1 Log Liver RAE 84 Figure 3.4 Continued. Effects of depleted liver RAE on spleen DC populations. A, myeloid DCs. B, lymphoid DCs. C, ratio of myeloid to lymphoid DCs. D, plasmacytoid DC 3. E, preDCs. Table insets indicate significant 0 coefficients, R2 values, and significance of model. 85 Percent Plasmacytoid DC (%) Plasmacytoid DC 0-45‘ [5 p value Liver RAE 0.08 0.40 0,40. Age -0.03 0.77 Gender 0.12 0.21 035. R2 = 0.02 0.57 0.30‘ 0.25 _ 0.20, 0.154 " _ " 0.10, 0.05- 0.00 . '04 w. h. .2 -1 0 1 Log Liver RAE 86 Figure 3.4 Continued. Effects of depleted liver RAE on spleen DC populations. A, myeloid DCs. B, lymphoid DCs. C, ratio of myeloid to lymphoid DCs. D, plasmacytoid DCs. E, preDCs. Table insets indicate significant [3 coefficients, R2 values, and significance of model. 87 Percent preDC (%) preDC 2.251 B p value 2 00- Liver RAE 0.10 0.31 ' Age -0.20 0.03 Gender 0.20 0.03 ”5‘ R2 = 0.09 0.02 1.501 1.25‘ 1.00- 0.751 0.50« 0.254 0.00 . . -2 -1 0 1 Log leer RAE 88 Figure 3.5. Effects of depleted liver RAE on spleen lymphocyte p0pulations. A, B cells. B, CD3+/CD4'/CD8' T cells. C, CD4 T cell. D, total CD8 T cells. E, Gr-lJ'/CD8+ memory T cells. Table insets indicate significant [3 coefficients, R2 values, and significance of model. 89 A B Lymphocytes 77.5- 75.0 72.5« 70.0« 1‘: 67.5« 3 650 8 '5 62 54 e. ' —— - _J m 60.0" § 57.5 3’ 550 ' I B p value 52.5- Liver RAE -0.00 0.98 Age 020 0.03 50'0' Gender 0.19 0.05 47.5« R2 = 0.08 0.03 45.0 I Y I .2 -1 0 1 Log Liver RAE 90 Figure 3.5 Continued. Effects of depleted liver RAE on spleen lymphocyte populations. A, B cells. B, CD3+/CD4'/CD8' T cells. C, CD4 T cell. D, total CD8 T cells. E, Gr-1+/CD8+ memory T cells. Table insets indicate significant [3 coefficients, R2 values, and significance of model. 91 Percent C03+CD4-CDBo (%) CD3+ICD4'ICDB' Lymphocytes [3 p value 13- Liver RAE 0.04 0.70 12‘ Age -0.18 0.06 Gender -0.1 1 0.25 1“ R2=O.O4 0.18 10. 9- -" 8. 7'1 6~ w , 5. 4. 3.. 2. 1 . 0 . -2 .1 1 Log Liver RAE 92 Figure 3.5 Continued. Effects of depleted liver RAE on spleen lymphocyte populations. A, B cells. B, CD3+/CD4°/CD8' T cells. C, CD4 T cell. D, total CD8 T cells. E, Gr-l+/CD8+ memory T cells. Table insets indicate significant [3 coefficients, R2 values, and significance of model. 93 Percent CD4 T cells (%) CD3+ICD4" T Lymphocytes -l j“ [3 p value Liver RAE 0.62 0.00 16+ Age 013 0.10 Gender 0.11 0.16 R2 = 0.36 0.00 .s j .s S" .s '5‘ 4.5.5.5 9'??? ‘P N4 0: -2 31 0 i Log Liver RAE 94 Figure 3.5 Continued. Effects of depleted liver RAE on spleen lymphocyte populations. A, B cells. B, CD3+/CD4’/CD8' T cells. C, CD4 T cell. D, total CD8 T cells. E, Gr-l+/CD8+ memory T cells. Table insets indicate significant [3 coefficients, R2 values, and significance of model. 95 Percent C08 T cells (%) CDT/COB” T Lymphocytes [3 p value ~ LiverRAE -0.07 0.44 Age 0.07 0.47 Gender -0.3 l 0.00 R2=0.10 0.01 31 I 1 Log Liver RAE 96 N4 #1 Figure 3.5 Continued. Effects of depleted liver RAE on spleen lymphocyte populations. A, B cells. B, CD3+/CD4'/CD8' T cells. C, CD4 T cell. D, total CD8 T cells. E, Gr-1+/CD8+ memory T cells. Table insets indicate significant [3 coefficients, R2 values. and significance of model. 97 CDB‘IGr-‘V Memory T Lymphocytes 7‘ S” E" ‘P w 1 J Percent Memory CD8 T cells (%) B p value 24 Liver RAE -0.62 0.00 Age 0.24 0.00 1. Gender -0.20 0.01 R2 = 0.38 0.00 O ' ' I ' I fl -2 -1 0 1 2 3 4 Log Liver RAE 98 Figure 3.6. Effects of depleted liver RAE on spleen PMN. Table inset indicates significant [3 coefficients, R2 values, and significance of model. 99 Neutrophils Percent PMN (%) 97 B p value Liver RAE —0.44 000 3‘ Age 0.14 0.10 Gender 0.06 0,51 71 R2 Z 0.20 0.00 6. 5. 4.1 3‘ '. 2‘ I I; 1. 0 . , . ‘ r _' '2 ‘1 ° 1 2 3 4 Log Liver RAE 100 CHAPTER 4: CONCLUSIONS & FUTURE DIRECTIONS Flow C ytometty of DCs The multicolor flow cytometry protocol developed can identify relevant DC populations of the spleen and Peyer’s patches of C57BL/6J mice (179). However, research continues to identify more DC subpopulations, such as CD103+ DCs present in the spleen and mucosal tissues capable of antigen cross-presentation, phagocytosis and presentation of apoptotic cells, and induction of tolerance (143, 159, 161). The BD Biosciences LSR II flow cytometer is capable of distinguishing and analyzing up to 18 separate fluorochromes. Therefore, open channels remain to add more monoclonal antibody-fluorochrome conjugates to identify more antigens on the DCs and therefore identify additional DC populations. However, careful antigen choices must be performed to ensure the antigen is specific to only one DC population or can be used in combination with other markers to distinguish one cell type. For example, in the murine spleen CD103)r DCs are a subpopulation of CD80L+ lymphoid DCs (161). Murine Model of Vitamin A Deficiency Others have reported methods of inducing vitamin A deficiency in mice using dietary restriction (91, 108, 120). The model employed in chapter 3 has characteristics of human vitamin A deficiency including depressed Th2 responses. We did not measure other parameters of deficiency, but the deficient animals likely suffer from impaired mucosal integrity and eventual protein-energy malnutrition if the mice were continued on the vitamin A-deficient diet. Weight, serum retinol, and liver retinol activity equivalent data provide further insight into other immunological studies of vitamin A-deficient mice. Dietary restriction of vitamin A leads to depleted serum retinol and liver retinyl-esters by 101 8 to 12 weeks of age (91). Continued restriction of vitamin A, over 8 to 12 weeks, leads to complications such as protein-energy malnutrition (PEM) due to inanition (1 12). Any animal that lost greater than or equal to 10% of total body weight was classified as suffering PEM and was excluded from our analysis. Therefore, we believe our immune cell population changes are attributable to VAD itself, and not related to PEM. The time course of analysis of the animals also demonstrates the effects of various degrees of vitamin A deficiency. DC Populations of Vitamin A-Deficient Mice Dietary depletion of vitamin A skews the proportion of DC populations in the spleen of mice. In accordance with our hypothesis, mice with the lowest liver retinol activity equivalents (RAE) had modestly decreased myeloid DCs and increased neutrophils. Unexpectedly, the lymphoid DCs were increased with more severe vitamin A deficiency. The increase of lymphoid DCs, responsible for stimulating Th1 responses in mice, is in accordance with reports that vitamin A deficiency increases Th1 responses at the expense of Th2 responses. The plasmacytoid and preDCs were unaffected by _ depletion of vitamin A. In summary, the main effect was increased lymphoid DCs which stimulate Th1 responses leading to cross-regulation of Th2 responses, but in combination with decreased myeloid DCs that stimulate Th2 responses. Other Immune Cell Populations of Vitamin A-Deficient Mice Vitamin A-deficient mice have skewed T lymphocyte populations, in addition to DC subpopulations. T helper CD4+ cells are decreased and memory CD8+ CTLs are increased in vitamin A-deficient animals compared to vitamin A-sufficient animals. The increased CTL population correlates with increased lymphoid DCs and Th1 responses of 102 vitamin A-deficient populations. B lymphocytes are unaffected by dietary depletion in mice. Other reports have shown vitamin A deficiency does not alter B lymphocyte numbers despite depressed Th2 antibody responses. In vivo, vitamin A deficiency has only minor affects on PMNs. Kuwata et al reported a marked significant increase in PMN populations in vitamin A-deficient SENCAR mice (108). However, the SENCAR mice were depleted of vitamin A for 14 weeks and therefore severely depleted of vitamin A and likely PEM. Glucocorticoid responses induced by PEM lead to increased PMN and decreased lymphocytes, both of which were observed in SENCAR vitamin A- deficient mice (181). Future Directions Vitamin A may have differential effects on other immune cell populations, DC populations, and cell populations of various tissues. Others have demonstrated the effects of vitamin A on various T cell populations. In vivo, we demonstrate the effects of vitamin A depletion on total CD4+, CD8+, memory CD8+, and CD3+/CD4'/CD8' T cell populations. However, there are other T lymphocyte populations dependent on vitamin A. As DC research continues to identify new DC populations, the effects of vitamin A deficiency on these populations can lead to more thorough understanding of the cooperation and coordination of the immune response in vitamin A-deficient populations. The effects of vitamin A are most dramatic in mucosal tissues, particularly the mucosal tissues of the gastrointestinal tract. The skewed cell numbers of various immune cell pOpulations is interesting, but the findings are limited in that we could not describe any functional changes in these studies. Therefore, various infection models could be employed in vitamin A—deficient animals to determine if the decreased cell populations 103 observed have the expected significant effects on the immune response to the various pathogens. Also it is important to determine the effects of supplementing vitamin A- deficient populations to determine if restoring adequate vitamin A status can return the immune cell populations to similar levels of vitamin A-sufficient animals and the kinetics of this restoration. Future directions therefore include expanding the research to include other immune cell populations and other secondary lymphoid tissues, determining DC function during vitamin A deficiency using infection models, and supplementing deficient animals to determine if the period of deficiency permanently depresses immune cell populations. We characterized many immune cell populations in these studies. However, Treg and Thl7 cells are also dependent on vitamin A (172, 174). In vitro T cells differentiate into Th17 cells in media lacking vitamin A. However, in media containing vitamin A the T cells differentiate into Treg cells (174). Thl7 cells are T lymphocytes that produce IL- 17 and have been shown to be responsible for development of autoimmune disease. Therefore, not only is vitamin A responsible for stimulating adequate Th2 antibody responses, but vitamin A may also be critical in maintaining tolerance to self antigens. CD103+ DCs are present in mucosal tissues and the germinal center marginal zones of the spleen and induce tolerance to self antigens (142, 143, 159, 161). Therefore, in combination with the Treg and Thl7 balance, a role of vitamin A in CD103+ DCs may provide insight into the observed imbalance of Treg and Thl7 cell populations in vitamin A-deficiency. Comparison of Treg, Th17 and CD103+ DCs of vitamin A-deficient to vitamin A-sufficient animals using similar in vivo methods would provide evidence of the role of vitamin A in development of tolerance. 104 It is of particular interest that lymphoid DCs and memory CD8+ T lymphocytes are both increased in vitamin A-deficient animals. Memory T cells, as well as natural killer (N K) and NKT cells, are maintained by the growth factor IL-15. Hepatic and pancreatic stellate cells, as well as DCs, produce IL-15 (185-187). Hepatic stellate cells and DCs can also store and metabolize vitamin A (135). Although vitamin A did not significantly alter IL-15 cytokine production by human T lymphocytes in vitro, the dual function of vitamin A metabolism and production of IL-15 by DCs and liver stellate cells leads to a possible inter-relationship that would be expected to selectively enhance memory T cell reliance during VAD (188). Therefore, the source of IL-15, plasmacytoid or lymphoid DCs, should be further investigated as well as any increase in lL-15 in VAD animals. Vitamin A has ubiquitous functions, but focused and major roles in the eye and mucosal tissues. The role of vitamin A in the eye employs the ll-cis-retinal form of vitamin A and not the all-trans-retinoic acid form functioning in the immune system. Therefore, characterization of DC populations and other immune cells of mucosal tissues in vitamin A-deficient animal may be significantly altered compared to vitamin A- sufficient populations. Homing of gut B and T lymphocytes and development of mucosal CD103+ DCs relies on the presence of vitamin A (135, 136, 143, 172, 189). Therefore, the cell populations may be decreased in the whole animal or in specific tissues due to the lack of homing responses. Since the spleen is a filtering organ for the entire body, our findings are representative of the immune system as a whole, but can not be extrapolated to specific body sites. The differences between vitamin A-sufficient and vitamin A- deficient animals may be more pronounced in the intestinal mucosa due to the added 105 homing function of vitamin A. In addition, the intestine is the first encounter immune cells have with dietary vitamin A and DCs possess the enzymes to convert vitamin A to the biologically active form. So in combination, the homing of immune cells, the enzymes to metabolize vitamin A, and the decreased immune cell populations observed in the spleen indicate that the intestinal mucosa is an important site to characterize immune cell populations of vitamin A-deficient animals. We show preliminary data for the effects of VAD of immune cells of the Peyer’s patches is Appendix II, but the data are unadjusted for age and gender and also lack statistical analysis. Skewed immune cell numbers of vitamin A-deficient animals does not directly indicate an increased susceptibility to infection or mortality from infection. Therefore, the vitamin A-de'ficient animals and vitamin A-sufficient control animals should be exposed to various pathogens eliciting various immune responses and self peptides leading to tolerance to determine if the changes in immune cell numbers leads to changes in disease susceptibility. A pathogen that leads to primarily a Th2 response is hypothesized to lead to increased susceptibility and mortality in vitamin A-deficient animals while a pathogen requiring primarily a Th1 response should survive the infection better than vitamin A-sufficient controls. Exposing the animals to a “self” antigen that leads to a Th17 autoimmune reaction is hypothesized to cause a more severe immune reaction in vitamin A—deficient animals compared to control, vitamin A—sufficient, animals. The models described could highlight the role of vitamin A, specifically the role of vitamin A in DCs, in clearing infections or establishing an autoimmune prone environment. Using the flow cytometry methodology described in chapter 2, a comprehensive overview of the immune response to various pathogen challenges in 106 vitamin A-deficient animals could establish a mechanism of vitamin A in the immune system during an immune challenge. The changes in immune cell numbers during vitamin A deficiency may be permanent changes. The deficient animals may not restore the immune cell numbers comparable to the sufficient animals even after supplementation. Animals depleted of vitamin A could be supplemented after various degrees of deficiency or after various times after deficiency has been established to determine appropriate supplementation strategies. Analysis of immune cell populations using the multicolor flow cytometry elucidated a role of vitamin A in maintaining DC subpopulations during homeostasis. However, more research needs to be conducted to further describe the increased susceptibility to infections in vitamin A-deficient populations. In addition, supplementation strategies may be improved by focusing on improved immune cell populations instead of circulating serum retinol levels. 107 APPENDIX I Supplemental data from the identification and enumeration of dendritic cell populations from individual mouse spleen and Peyer’s patches using flow cytometric analysis (Chapter 2). 108 Supplemental Figure 1.1. LSR II optics block and filter scheme of the fluorescent channels utilized for data collection. 109 Blue Laser (488 nm) PerCP-Cy5.5 pl Red Laser (633 nm) APC-Cy7 APC Alexa Fluor 700 110 Supplemental Figure 1.2. Comparison of compensated and uncompensated data. Left panels (A, C, E, G) are compensated contour plots. Right panels (B, D, F, H) are uncompensated contour plots. Percentage of compensation employed, calculated from CompBead fluorescence matrix from the automatic compensation feature of the DIVA software, is represented above the compensated panel. 111 > MHC ll-FlTC-A Gr1 PerCP-Cy5.5-A 8220 APC-A Gr1 PerCP-CyS.S-A 18.04% CDllc PE-A 17.39% C083 PE-Cy7-A 18.15% c0110 APC-Cy7-A 13.02% CDllc PE-A MHC ll-FlTC-A Gr1 PerCP-Cy5.5-A 8220 APC-A Gr1 PerCP-Cy5.5-A r I .7639 CDllc PE-A C083 PE-CV7-A 0} CDl 1b APC-CV7-A CDllc PE-A 112 Supplemental Figure 1.3. Flow diagram of Boolean logic used in sequential gating of spleen cell populations. Final populations gated and enumerated are shown in red boxes. 113 Ow 259$ gnu-mmiooo u..- $903300 no «OE 53:00 agate—22.6 .o 2:88.: 0.354198% ..§§8.2=8:.5.ozse§aa.oso “=95. . .SBEEJSoozoifaonngsc ages. 9...: E 2.3... 4110...... .3 2.. age 2.2... 8 =. afién 3 58:8 92.88 as. 533:8 .o... s 8... 58:8 E E m. 2:9“. Or 239m :54? 5808 En! §o§8§§u.88:.6.oz:$8=2§:!8o§ o 0:23 2: 38 h 382:3 u..- 23 so .5823 5.3.2... lg u. 2.5... £288.: 2.3 r 114 Supplemental Table 1.1. BD® Biosciences LSR II flow cytometer laser parameters used in flow cytometry data collection. Parameters (Channel) Voltage Scale FSC 415 Linear SSC 382 Linear F ITC (FL 1 ) 484 Bi-exponential PE (FL2) 415 Bi-exponential PerCP-Cy5.5 (F L3) 725 Bi-exponential PE-Cy7 (FL4) 525 ‘ Bi-exponential APC (F L5) 516 Bi-exponential APC-Cy7 (FL6) 535 Bi-exponential 115 Supplemental Table 1.2. BD® Biosciences LSR 11 general compensation matrix. F luorochromes Value (%) PE - FITC 18.04 PeGC—Cy5.5 - FITC 1.91 PE-Cy7 — FITC 0.19 APC — FITC 0.28 APC-Cy7 — FITC 0.00 FITC — PE 0.46 PeGC-Cy5.5 — PE 13.02 PE-Cy7 — PE 1.26 APC — PE 0.10 APC-Cy7 — PE 0.00 FITC -— PeGC-Cy5.5 0.00 PE — PeGC-Cy5.5 0.00 PE-Cy7 — PeGC-Cy5.5 17.39 APC — PeGC-Cy5.5 1.94 APC-Cy7 — PeGC-Cy5.5 1.67 _ FITC — PE-Cy7 0.16 ~ PE — PE-Cy7 3.27 K PeGC-Cy5.5 — Pe-Cy7 0.84 116 Supplemental Table 1.2 Continued. BD® Biosciences LSR II general compensation matrix. APC — PE—Cy7 0.14 APC-Cy7 — PE-Cy7 2.24 FITC —- APC 0.00 PE — APC 0.00 PerCP-Cy5.5 — APC 0.84 PE-Cy7 -— APC 0.14 APC-Cy7 — APC 3.09 FITC — APC-Cy7 0.00 PE — APC-Cy7 0.00 PeGC-Cy5.5 — APC-Cy7 0.11 PE-Cy7 —— APC-Cy7 4.37 APC — APC-Cy7 18.15 117 Preliminary data on the effects of vitamin A deficiency on immune cells of the Peyer ’s patch es. This data is not adjusted for gender or age effects, and has not been statistically analyzed. APPENDIX 11 In addition this data has not been adjusted for total cell numbers per Peyer’s patch to decrease variability of numbers of Peyer’s patches excised from each animal. Supplemental Figure 2.1. Immune cell percentages of vitamin A-deficient mice. A Percent (%) Percent (%) Myeloi d y = 0.0234ln(x) + 0.7579 R2 = 0.0107 2.50 2.00 .0- 1 50 ’ ’ . 9 j .. . O: .. O 1.00 ’ 43* ’ 41:...‘5— —_ O O 0.50 .f ' t o o .0 Q 9 O O 0.00 1— I 1 4 1 A. 1 1 0.01 0.1 1 10 100 1000 Log Liver RAE . y = 0.00281n(x) + 0.1545 Lympho'd R2 = 0.0083 0.5 0.4 ’ O 0.3 o 9 . 0-2 M 0’. Q 0 O a L 0.11 ~f .fi . O l l l l l 0.01 0.1 1 10 100 1000 Log Liver RAE 118 Percent (%) Percent (%) Plasmacytoid y = O°08()71r1(><) + 0.4428 R2 = 0.084 3.5 3 3— 2.5 2 1.5 O . O 1 ‘ . r 0-5 —— W M Q 0 r , ,‘ ’ 0 o T . 0.01 0.1 1 10 100 Log Liver RAE y = 0.037ln(x) + 0.0628 preDC R2 = 0.1001 1.2 1 ._ 0 0.8 . . . 0.6 L, . o 0.4 + o /9.5 0.2 o O ‘ ' . W 0 . I -0200] D 1 10 100 JflOO Log Liver RAE 119 Percent (%) '11 Percent (%) y = -0.0111n(x) + 0.4129 Other DCs R2 = 0.0062 1.2 1 O O 0.8 ‘ W O 0.6 O . ‘ 0.4 . 0.2 . ;+ 99‘ ’ ”co 8 0 r r A; Q 1 1 0.01 0.1 1 10 100 Log Liver RAE y = 0.58411n(x) + 17.825 75 T cells R2 - 00545 35 30 25 L #9 o o 0‘ O O 20 f /M 15 . ' I ‘~ . 10 W + + S 0 l I V I 1 0.01 0.1 l 10 100 1000 Log Liver RAE 120 1:: Percent (%) Percent (%) Or-‘NUJAU’I CD4 T cells y = 0.2011110.) + 6-8741 R2 = 0.1138 10 o 8 0 o 3 NO . O . O. 6 . . . 4 9 9 0‘ g ’ o 2 O I I . 0.01 1 10 100 1000 Log Liver RAE y = -0.059ln(x) + 25571 CD8 T cells R2 = 0.0306 0.01 10 100 1000 Log Liver RAE 121 10. 11. 12. LITERATURE CITED Janeway, C. 2005. Immunobiology .' the immune system in health and disease. Garland Science, New York. Steinman, R. M., and Z. A. Cohn. 1973. Identification of a novel cell type in peripheral lymphoid organs of mice. 1. Morphology, quantitation, tissue distribution. J Exp Med 137:1142-1162. Steinman, R. M., G. Kaplan, M. D. Witmer, and Z. A. Cohn. 1979. Identification of a novel cell type in peripheral lymphoid organs of mice. V. Purification of spleen dendritic cells, new surface markers, and maintenance in vitro. J Exp Med 149:1-16. D'Amico, A., and L. Wu. 2003. The early progenitors of mouse dendritic cells and plasmacytoid predendritic cells are within the bone marrow hemopoietic precursors expressing Flt3. J Exp Med 198:293-303. Shortman, K., and S. H. Naik. 2007. Steady-state and inflammatory dendritic-cell development. Nat Rev Immunol 7: 19-30. Kabashima, K., T. A. Banks, K. M. Ansel, T. T. Lu, C. F. Ware, and J. G. Cyster. 2005. Intrinsic lymphotoxin-beta receptor requirement for homeostasis of lymphoid tissue dendritic cells. Immunity 22:439-450. lnaba, K., W. J. Swiggard, R. M. Steinman, N. Romani, G. Schuler, and C. Brinster. 2009. Isolation of dendritic cells. Curr Protoc Immunol Chapter 3:Unit 3 7. Shortman, K., and Y. J. Liu. 2002. Mouse and human dendritic cell subtypes. Nat Rev Immunol 2:151-161. Ardavin, C. 2003. Origin, precursors and differentiation of mouse dendritic cells. Nat Rev Immunol 3:582-590. Maldonado-Lopez, R., T. De Smedt, P. Michel, J. Godfroid, B. Pajak, C. Heirman, K. Thielemans, 0. Leo, J. Urbain, and M. Moser. 1999. CD8alpha+ and CD8alpha- subclasses of dendritic cells direct the development of distinct T helper cells in vivo. J Exp Med 189:587-592. Pulendran, B., J. 11.. Smith, G. Caspary, K. Brasel, D. Pettit, E. Maraskovsky, and C. R. Maliszewski. 1999. Distinct dendritic cell subsets differentially regulate the class of immune response in vivo. Proc Natl Acad Sci U S A 96:1036—1041. Nakano, H., M. Yanagita, and M. D. Gunn. 2001. CD11c(+)B220(+)Gr—1(+) cells in mouse lymph nodes and spleen display characteristics of plasmacyto1d dendritic cells. JExp Med 194:1171-1178. 122 l3. 14. 15. 16. 17. 18. 19. 20. 21. 22. 23. Martin, P., G. M. Del Hoyo, F. Anjuere, C. F. Arias, H. H. Vargas, L. A. Fernandez, V. Parrillas, and C. Ardavin. 2002. Characterization of a new subpopulation of mouse CD8alpha+ 8220+ dendritic cells endowed with type 1 interferon production capacity and tolerogenic potential. Blood 1002383-390. Liu, Y. J. 2005. IPC: professional type 1 interferon-producing cells and plasmacytoid dendritic cell precursors. Annu Rev Immunol 23:275-306. Hochrein, H., M. O'Keeffe, and H. Wagner. 2002. Human and mouse plasmacytoid dendritic cells. Hum Immunol 63:1 103-1 1 10. Nikolic, T., G. M. Dingjan, P. J. Leenen, and R. W. Hendriks. 2002. A subfraction of 8220(+) cells in murine bone marrow and spleen does not belong to the B cell lineage but has dendritic cell characteristics. Eur J Immunol 32:686- 692. Vremec, D., M. Zorbas, R. Scollay, D. J. Saunders, C. F. Ardavin, L. Wu, and K. Shortman. 1992. The surface phenotype of dendritic cells purified from mouse thymus and spleen: investigation of the CD8 expression by a subpopulation of dendritic cells. J Exp Med 176:47-58. Vremec, D., J. Pooley, H. Hochrein, L. Wu, and K. Shortman. 2000. CD4 and CD8 expression by dendritic cell subtypes in mouse thymus and spleen. J Immunol 164:2978-2986. Kamath, A. T., J. Pooley, M. A. O’Keeffe, D. Vremec, Y. Zhan, A. M. Lew, A. D'Amico, L. Wu, D. F. Tough, and K. Shortman. 2000. The development, maturation, and turnover rate of mouse spleen dendritic cell pOpulations. J Immunol 16526762-6770. Steinman, R. M., L. Bonifaz, S. Fujii, K. Liu, D. Bonnyay, S. Yamazaki, M. Pack, D. Hawiger, T. lyoda, 1K. lnaba, and M. C. Nussenzweig. 2005. The innate functions of dendritic cells in peripheral lymphoid tissues. Adv Exp Med Biol 560:83-97. Diao, J., E. Winter, C. Cantin, W. Chen, L. Xu, D. Kelvin, J. Phillips, and M. S. Cattral. 2006. In situ replication of immediate dendritic cell (DC) precursors contributes to conventional DC homeostasis in lymphoid tissue. J Immunol 17627196-7206. del Hoyo, G. M., P. Martin, H. H. Vargas, S. Ruiz, C. F. Arias, and C. Ardavin. 2002. Characterization of a common precursor population for dendritic cells. Nature 415:1043-1047. Luft, T., M. Jefford, P. Luetjens, T. Toy, H. Hochrein, K. A. Mastermari, C. Maliszewski, K. Shortman, J. Cebon, and E. Maraskovsky. 2002._Funct10nally distinct dendritic cell (DC) populations induced by phys1olog1c stlmuh: 123 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. prostaglandin E(2) regulates the migratory capacity of specific DC subsets. Blood 100:1362-1372. Kamath, A. T., S. Henri, F. Battye, D. F. Tough, and K. Shortman. 2002. Developmental kinetics and lifespan of dendritic cells in mouse lymphoid organs. Blood 100:1734-1741. Salomon, B., J. L. Cohen, C. Masurier, and D. Klatzmann. 1998. Three populations of mouse lymph node dendritic cells with different origins and dynamics. J Immunol 160:708-717. Leenen, P. J ., K. Radosevic, J. S. Voerman, B. Salomon, N. van Rooijen, D. Klatzmann, and W. van Ewijk. 1998. Heterogeneity of mouse spleen dendritic cells: in vivo phagocytic activity, expression of macrophage markers, and subpopulation turnover. J Immunol 160:2166-2173. Leon, B., G. Martinez del Hoyo, V. Parrillas, H. H. Vargas, P. Sanchez-Mateos, N. Longo, M. Lopez-Bravo, and C. Ardavin. 2004. Dendritic cell differentiation potential of mouse monocytes: monocytes represent immediate precursors of CD8- and CD8+ splenic dendritic cells. Blood 103:2668-2676. Rissoan, M. C., V. Soumelis, N. Kadowaki, G. Grouard, F. Briere, R. de Waal Malefyt, and Y. J. Liu. 1999. Reciprocal control of T helper cell and dendritic cell differentiation. Science 283:1 183-1186. Harrison, E. H. 2005. Mechanisms of digestion and absorption of dietary vitamin A. Annu Rev Nutr 25:87-103. Lindqvist, A., and S. Andersson. 2004. Cell type-specific expression of beta- carotene 15,15'-mono-oxygenase in human tissues. J Histochem C ytochem 52:491-499. Parker, R. S. 1996. Absorption, metabolism, and transport of carotenoids. Faseb J 10:542-551. Rigtrup. K. M., and D. E. Ong. 1992. A retinyl ester hydrolase activity intrinsic to the brush border membrane of rat small intestine. Biochemistry 31 :2920-2926. Rigtrup, K. M., B. Kakkad, and D. E. Ong. 1994. Purification and partial characterization of a retinyl ester hydrolase from the brush border of rat small intestine mucosa: probable identity with brush border phospholipase B. Biochemistry 33 :2661 —2666. Erlanson, C., and B. Borgstrom. 1968. The identity of vitamin A esterase activity of rat pancreatic juice. Biochim Biophys Acta 167:629-631. 124 35. 36. 37. 38. 39. 40. 41. 42. 43. 44. 45. 46. 47. Blomhoff, R., M. H. Green, J. B. Green, T. Berg, and K. R. Norum. 1991. Vitamin A metabolism: new perspectives on absorption, transport, and storage. Physiol Rev 71:951-990. Blomhoff, R., M. H. Green, T. Berg, and K. R. Norum. 1990. Transport and storage of vitamin A. Science 250:399-404. Goodman, D. S., R. Blomstrand, B. Werner, H. S. Huang, and T. Shiratori. 1966. The intestinal absorption and metabolism of vitamin A and beta-carotene in man. J Clin Invest 45:1615-1623. Paik, J ., S. Vogel, L. Quadro, R. Piantedosi, M. Gottesman, K. Lai, L. Hamberger, M. Vieira Mde, and W. S. Blaner. 2004. Vitamin A: overlapping delivery pathways to tissues from the circulation. J Nutr 134:276S-28OS. Wake, K. 1974. Development of vitamin A-rich lipid droplets in multivesicular bodies of rat liver stellate cells. J Cell Biol 63:683-691. Monaco, H. L. 2000. The transthyretin-retinol-binding protein complex. Biochim Biophys Acta 1482:65-72. Soprano, D. R., C. B. Pickett, J. E. Smith, and D. S. Goodman. 1981. Biosynthesis of plasma retinol-binding protein in liver as a larger molecular weight precursor. J Biol Chem 256:8256-8258. Kanai, M., A. Raz, and D. S. Goodman. 1968. Retinol-binding protein: the transport protein for vitamin A in human plasma. J C [in Invest 47:2025-2044. Stephensen, C. B., and G. Gildengorin. 2000. Serum retinol, the acute phase response, and the apparent misclassification of vitamin A status in the third National Health and Nutrition Examination Survey. Am J Clin Nutr 72:1170- 1178. Kawaguchi, R., J. Yu, J. Honda, J. Hu, J. Whitelegge, P. Ping, P. Wiita, D. Bok, and H. Sun. 2007. A membrane receptor for retinol binding protein mediates cellular uptake of vitamin A. Science 315:820-825. Duester, G. 2000. Families of retinoid dehydrogenases regulating vitamin A function: production of visual pigment and retinoic acid. Eur J Biochem 267:4315-4324. Bashor, M. M., D. O. Toft, and F. Chytil. 1973. In vitro binding of retinol to rat- tissue components. Proc Natl Acad Sci U S A 70:3483-3487. Goodman, D. W., H. S. Huang, and T. Shiratori. 1965. Tissue Distribution and Metabolism of Newly Absorbed Vitamin a in the Rat. J Lipid Res 62390-396. 125 48. 49. 50. 51. 52. 53. 54. 55. 56. 57. 58. 59. 60. Tsutsumi, C., M. Okuno, L. Tannous, R. Piantedosi, M. Allan, D. S. Goodman, and W. S. Blaner. 1992. Retinoids and retinoid-binding protein expression in rat adipocytes. J Biol Chem 267: 1805-1 8 10. Hicks, V. A., D. B. Gunning, and J. A. Olson. 1984. Metabolism, plasma transport and biliary excretion of radioactive vitamin A and its metabolites as a function of liver reserves of vitamin A in the rat. J Nutr 114:1327-1333. Stephensen, C. B., J. O. Alvarez, J. Kohatsu, R. Hardmeier, J. 1. Kennedy, Jr., and R. B. Gammon, Jr. 1994. Vitamin A is excreted in the urine during acute infection. Am J Clin Nutr 60:388-392. Cullum, M. E., and M. H. Zile. 1985. Metabolism of all-trans-retinoic acid and all-trans-retinyl acetate. Demonstration of common physiological metabolites in rat small intestinal mucosa and circulation. J Biol Chem 260:10590-10596. Marino, M., D. Andrews, D. Brown, and R. T. McCluskey. 2001. Transcytosis of retinol-binding protein across renal proximal tubule cells after megalin (gp 330)- mediated endocytosis. J Am Soc Nephrol 12:63 7-648. Soprano, D. R., K. J. Soprano, and D. S. Goodman. 1986. Retinol-binding protein messenger RNA levels in the liver and in extrahepatic tissues of the rat. J Lipid Res 27:166-171. Clagett—Dame, M., and H. F. DeLuca. 2002. The role of vitamin A in mammalian reproduction and embryonic development. Annu Rev Nutr 22:347-381. Mathies, R., and L. Stryer. 1976. Retinal has a highly dipolar vertically excited singlet state: implications for vision. Proc Natl Acad Sci U S A 73:2169-2173. Petkovich, M., N. J. Brand, A. Krust, and P. Chambon. 1987. A human retinoic acid receptor which belongs to the family of nuclear receptors. Nature 330:444- 450. Giguere, V., E. S. Ong, P. Segui, and R. M. Evans. 1987. Identification ofa receptor for the morphogen retinoic acid. Nature 330:624-629. Klein, E. S., J. W. Wang, B. Khalifa, S. A. Gavigan, and R. A. Chandraratna. 2000. Recruitment of nuclear receptor corepressor and coactivator to the retinoic acid receptor by retinoid ligands. Influence of DNA-heterodimer interactions. J Biol Chem 275:19401-19408. Chambon, P. 1996. A decade of molecular biology of retinoic acid receptors. Faseb J 10:940-954. Bastien, J ., and C. Rochette-Egly. 2004. Nuclear retinoid receptors and the transcription of retinoid-target genes. Gene 328:1-16. 126 61. 62. 63. 64. 65. 66. 67. 68. 69. 70. 71. 72. 73. 74. Bernhardt, I. B., and D. J. Dorsey. 1974. Hypervitaminosis A and congenital renal anomalies in a human infant. Obstet Gynecol 43:750-755. Rothman, K. J., L. L. Moore, M. R. Singer, U. S. Nguyen, S. Mannino, and A. Milunsky. 1995. Teratogenicity of high vitamin A intake. N Engl J Med 333:1369—1373. Zile, M. H. 2001. Function of vitamin A in vertebrate embryonic development. J Nutr 131:705-708. Thompson, J. N., J. M. Howell, and G. A. Pitt. 1964. Vitamin a and Reproduction in Rats. Proc R Soc Lond B Biol Sci 159:510-535. Wellik, D. M., and H. F. DeLuca. 1995. Retinol in addition to retinoic acid is required for successful gestation in vitamin A-deficient rats. Biol Reprod 53:1392-1397. Wald, G. 1968. The molecular basis of visual excitation. Nature 219:800-807. Yoshizawa, T., and G. Wald. 1963. Pre-lumirhodopsin and the bleaching of visual pigments. Nature 19721279-1286. Schoenlein, R. W., L. A. Peteanu, R. A. Mathies, and C. V. Shank. 1991. The first step in vision: femtosecond isomerization of rhodopsin. Science 254:412-415. Mata, N. L., and A. T. Tsin. 1998. Distribution of l l-cis LRAT, 11-cis RD and 11-cis REH in bovine retinal pigment epithelium membranes. Biochim Biophys Acta 1394:16-22. Nau, H., W. S. Blaner, and A. Agadir. 1999. Retinoids : the biochemical and molecular basis of vitamin A and retinoid action. Springer, Berlin ; New York. Balmer, J. E., and R. Blomhoff. 2002. Gene expression regulation by retinoic acid. JLipid Res 43:1773-1808. Umesono, K., and R. M. Evans. 1989. Determinants of target gene specificity for steroid/thyroid hormone receptors. Cell 57:1139-1 146. Kato, S., H. Sasaki, M. Suzawa, S. Masushige, L. Tora, P. Chambon, and H. Gronemeyer. 1995. Widely spaced, directly repeated PuGGTCA elements act as promiscuous enhancers for different classes of nuclear receptors. Mol Cell Biol 15:5858-5867. Kakizuka, A., W. H. Miller, Jr., K. Umesono, R. P. Warrell, Jr., S. R. Frankel, V. V. Murty, E. Dmitrovsky, and R. M. Evans. 1991. Chromosomal translocation t(15;17) in human acute promyelocytic leukemia fuses RAR alpha with a novel putative transcription factor, PML. Cell 66:663-674. 127 75. 76. 77. 78. 79. 80. 81. 82. 83. 84. 85. 86. Frankel, S. R., A. Eardley, G. Heller, E. Berman, W. H. Miller, Jr., E. Dmitrovsky, and R. P. Warrell, Jr. 1994. All-trans retinoic acid for acute promyelocytic leukemia. Results of the New York Study. Ann Intern Med 120:278-286. West, K. P., Jr. 2002. Extent of vitamin A deficiency among preschool children and women of reproductive age. J Nutr 132:2857S-2866S. , Pilch, S. M. 1987. Analysis of vitamin A data from the health and nutrition examination surveys. J Nutr 117:636-640. Loevinsohn, B. P., R. W. Sutter, and M. O. Costales. 1997. Using cost- effectiveness analysis to evaluate targeting strategies: the case of vitamin A supplementation. Health Policy Plan 12:29-37. Ye, X., S. Al—Babili, A. Kloti, J. Zhang, P. Lucca, P. Beyer, and I. Potrykus. 2000. Engineering the provitarnin A (beta-carotene) biosynthetic pathway into (carotenoid-free) rice endosperm. Science 287:303-305. Humphrey, J. H., K. P. West, Jr., and A. Sommer. 1992. Vitamin A deficiency and attributable mortality among under-S-year-olds. Bull World Health Organ 70:225-232. Fawzi, W. W., T. C. Chalmers, M. G. Herrera, and F. Mosteller. 1993. Vitamin A supplementation and child mortality. A meta-analysis. Jama 269:898-903. Shils, M. E., and M. Shike. 2006. Modern nutrition in health and disease. Lippincott Williams & Wilkins, Philadelphia. Rojanapo, W., J. A. Olson, and A. J. Lamb. 1980. Biochemical and immunological characterization and the synthesis of rat intestinal glycoproteins following the induction of rapid synchronous vitamin A deficiency. Biochim Biophys Acta 633:386-399. Olson, J. A., W. Rojanapo, and A. J. Lamb. 1981. The effect of vitamin A status on the differentiation and function of goblet cells in the rat intestine. Ann N Y AcadSci 359:181-191. Kligman, L. H., and A. M. Kligman. 1979. The effect on rhino mouse skin of agents which influence keratinization and exfoliation. J Invest Dermatol 73:354- 358. Goldfarb, R. H., and R. B. Herberman. 1981. Natural killer cell reactivity: regulatory interactions and among phorbol ester, interferon, cholera toxm, and retinoic acid. J Immunol 12622129-2135. 128 87. 88. 89. 90. 91. 92. 93. 94. 95. 96. 97. 98. 99. Santoni, A., S. Cerruti Sola, M. Giovarelli, P. Martinetto, D. Vietti, and G. Forni. 1986. Modulation of natural killer activity in mice by prolonged administration of various doses of dietary retinoids. Nat Immun Cell Growth Regul 5:259-266. Goettsch, W., Y. Hatori, and R. P. Shanna. 1992. Adjuvant activity of all-trans- retinoic acid in C57B1/6 mice. Int J Immunopharmacol 14:143-150. Ross, A. C. 1996. Vitamin A deficiency and retinoid repletion regulate the antibody response to bacterial antigens and the maintenance of natural killer cells. C [in Immunol Immunopathol 80:863-72. Zhao, 2., D. M. Murasko, and A. C. Ross. 1994. The role of vitamin A in natural killer cell cytotoxicity, number and activation in the rat. Nat Immun 13:29-41. smith, S. M., N. S. Levy, and C. E. Hayes. 1987. Impaired immunity in vitamin A-deficient mice. J Nutr 117:857-865. Tachibana, K., S. Sone, E. Tsubura, and Y. Kishino. 1984. Stimulatory effect of vitamin A on tumoricidal activity of rat alveolar macrophages. Br J Cancer 49:343-348. Moriguchi, S., L. Werner, and R. R. Watson. 1985. High dietary vitamin A (retinyl palmitate) and cellular immune functions in mice. Immunology 56:169- 177. Hatchigian, E. A., J. I. Santos, S. A. Broitman, and J. J. Vitale. 1989. Vitamin A supplementation improves macrophage function and bacterial clearance during experimental salmonella infection. Proc Soc Exp Biol Med 191247-54. Crowle, A. J ., and E. J. Ross. 1989. Inhibition by retinoic acid of multiplication of virulent tubercle bacilli in cultured human macrophages. Infect Immun 57:840- 844. Rhodes, J., and S. Oliver. 1980. Retinoids as regulators of macrophage function. Immunology 40:467-472. Cantoma, M. T., F. E. Nashold, and C. E. Hayes. 1995. Vitamin A deficiency results in a priming environment conducive for Th1 cell development. Eur J Immunol 25: 1673-1679. Na, S. Y., B. Y. Kang, S. W. Chung, S. J. Han, X. Ma, G. Trinchieri, S. Y. Im, J. W. Lee, and T. S. Kim. 1999. Retinoids inhibit interleukin-12 production in macrophages through physical associations of retinoid X receptor and NFkappaB. J Biol Chem 274:7674-7680. Wiederrnann, U., X. J. Chen, L. Enerback, L. A. Hanson, H. Kahu, and U. I. Dahlgren. 1996. Vitamin A deficiency increases inflammatory responses. Scand J Immunol 44:578-584. 129 100. 101. 102. 103. 104. 105. 106. 107. 108. 109. 1110. 111. 112. Shams, N. B., C. V. Reddy, K. Watanabe, S. A. Elgebaly, L. A. Hanninen, and K. R. Kenyon. 1994. Increased interleukin-1 activity in the injured vitamin A- deficient cornea. Cornea 13: 156-166. Aukrust, P., F. Muller, T. Ueland, A. M. Svardal, R. K. Berge, and S. S. F roland. 2000. Decreased vitamin A levels in common variable immunodeficiency: vitamin A supplementation in vivo enhances immunoglobulin production and downregulates inflammatory responses. Eur J C [in Invest 30:252-259. elSisi, A. E., D. L. Earnest, and I. G. Sipes. 1993. Vitamin A potentiation of carbon tetrachloride hepatotoxicity: role of liver macrophages and active oxygen species. Toxicol Appl Pharmacol 119:295-301. Nauss, K. M., C. A. Anderson, M. W. Conner, and P. M. Newberne. 1985. Ocular infection with herpes simplex virus (HSV-l) in vitamin A-deficient and control rats. JNutr 115:1300-1315. Wiedermann, U., A. Tarkowski, T. Bremell, L. A. Hanson, H. Kahu, and U. I. Dahlgren. 1996. Vitamin A deficiency predisposes to Staphylococcus aureus infection. Infect Immun 64:209-214. Sijtsma, S. R., J. H. Rombout, M. J. Dohmen, C. E. West, and A. J. van der Zijpp. 1991. Effect of vitamin A deficiency on the activity of macrophages in Newcastle disease virus-infected chickens. Vet Immunol Immunopathol 28:17-27. Nauss, K. M., D. A. Mark, and R. M. Suskind. 1979. The effect of vitamin A deficiency on the in vitro cellular immune response of rats. J Nutr 109:1815-1823. Zhao, Z., and A. C. Ross. 1995. Retinoic acid repletion restores the number of leukocytes and their subsets and stimulates natural cytotoxicity in vitamin A- deficient rats. J Nutr 125:2064-2073. Kuwata, T., I. M. Wang, T. Tamura, R. M. Ponnamperuma, R. Levine, K. L. Holmes, H. C. Morse, L. M. De Luca, and K. Ozato. 2000. Vitamin A deficiency in mice causes a systemic expansion of myeloid cells. Blood 95:3349-3356. Ongsakul, M., S. Sirisinha, and A. J. Lamb. 1985. Impaired blood clearance of bacteria and phagocytic activity in vitamin A-deficient rats. Proc Soc Exp Biol Med 178:204-208. Twining, S. S., D. P. Schulte, P. M. Wilson, B. L. Fish, and J. E. Moulder. 1997. Vitamin A deficiency alters rat neutrophil function. J Nutr 127:558-565. Collins, S. J. 2002. The role of retinoids and retinoic acid receptors in normal hematOpoiesis. Leukemia 16:1896-1905. Stephensen, C. B. 2001. Vitamin A, infection, and immune function. Annu Rev Nutr 21:167-192. 130 113. 114. 115. 116. 117. 118. 119. 120. 121. 122. 123. 124. 125. Jayalakshmi, V. T., and C. Gopalan. 1958. Nutrition and tuberculosis. 1. An epidemiological study. Indian J Med Res 46:87-92. Wiedermann, U., L. A. Hanson, H. Kahu, and U. I. Dahlgren. 1993. Aberrant T- cell function in vitro and impaired T-cell dependent antibody response in vivo in vitamin A-deficient rats. Immunology 80:581-586. Spom, M. B., A. B. Roberts, and D. S. Goodman. 1994. The Retinoids .' biology. chemistry, and medicine. Raven Press, New York. Sidell, N., and F. Ramsdell. 1988. Retinoic acid upregulates interleukin-2 receptors on activated human thymocytes. Cell Immunol 115:299-309. Sidell, N., U. Kummer, D. Aframian, and S. Thierfelder. 1997. Retinoid regulation of interleukin-2 receptors on human T-cells. Cell Immunol 179:1 16- 125. Sidell, N., B. Chang, and L. Bhatti. 1993. Upregulation by retinoic acid of interleukin-Z-receptor mRNA in human T lymphocytes. Cell Immunol 146:28-37. Fegan, C., R. Bailey-Wood, S. Coleman, S. A. Phillips, L. Neale, T. Hoy, and J. A. Whittaker. 1995. All trans retinoic acid enhances human LAK activity. Eur J Haematol 54:95-100. Stephensen, C. B., Z. Moldoveanu, and N. N. Gangopadhyay. 1996. Vitamin A deficiency diminishes the salivary immunoglobulin A response and enhances the serum immunoglobulin G response to influenza A virus infection in BALB/c mice. J Nutr 126:94-102. Cui, D., Z. Moldoveanu, and C. B. Stephensen. 2000. High-level dietary vitamin A enhances T-helper type 2 cytokine production and secretory immunoglobulin A response to influenza A virus infection in BALB/c mice. J Nutr 130:1132-1139. Cantoma, M. T., F. E. Nashold, and C. B. Hayes. 1994. In vitamin A deficiency multiple mechanisms establish a regulatory T helper cell imbalance with excess Th1 and insufficient Th2 function. J Immunol 152: 1515-1522. Smith, S. M., and C. E. Hayes. 1987. Contrasting impairments in IgM and IgG responses of vitamin A-deficient mice. Proc Natl Acad Sci U S A 84:5878-5882. Wiedermann, U., L. A. Hanson, J. Holmgren, H. Kahu, and U. I. Dahlgren. 1993. Impaired mucosal antibody response to cholera toxin in vitamin A-deficient rats immunized with oral cholera vaccine. Infect Immun 61 :3952-3957. Semba, R. D., Muhilal, A. L. Scott, G. Natadisastra, S. Wirasasmita, L. Mele, E. Ridwan, K. P. West, Jr., and A. Sommer. 1992. Depressed immune response to tetanus in children with vitamin A deficiency. J Nutr 122:101-107. 131 126. 127. 128. I29. 130. 131. 132. 133. 134. 135. 136. 137. Palmer, S. 1978. Influence of vitamin A nutriture on the immune response: findings in children with Down's syndrome. Int J Vitam Nutr Res 48: 188-216. Karalliedde, S., S. Dissanayake, and T. W. Wikramanayake. 1979. Salivary immunoglobulin A in vitamin A deficiency. Ceylon Med J 24:69-70. Sirisinha, S., M. D. Darip, P. Moongkamdi, M. Ongsakul, and A. J. Lamb. 1980. Impaired local immune response in vitamin A-deficient rats. C [in Exp Immunol 40:127-135. Stephensen, C. B., S. R. Blount, T. R. Schoeb, and J. Y. Park. 1993. Vitamin A deficiency impairs some aspects of the host response to influenza A virus infection in BALB/c mice. J Nutr 123:823-833. Gangopadhyay, N. N., Z. Moldoveanu, and C. B. Stephensen. 1996. Vitamin A deficiency has different effects on immunoglobulin A production and transport during influenza A infection in BALB/c mice. J Nutr 126:2960-2967. Carman, J. A., S. M. Smith, and C. B. Hayes. 1989. Characterization of a helper T lymphocyte defect in vitamin A-deficient mice. J Immunol 142:388-393. Racke, M. K., D. Burnett, S. H. Pak, P. S. Albert, B. Carmella, C. S. Raine, D. E. McFarIin, and D. E. Scott. 1995. Retinoid treatment of experimental allergic encephalomyelitis. IL-4 production correlates with improved disease course. J Immunol 154:450-458. Hengesbach, L. M., and K. A. Hoag. 2004. Physiological concentrations of retinoic acid favor myeloid dendritic cell development over granulocyte development in cultures of bone marrow cells from mice. J Nutr 134:2653-2659. Douer, D., L. Ramezani, J. Parker, and A. M. Levine. 2000. All-trans-retinoic acid effects the growth, differentiation and apoptosis of normal human myeloid progenitors derived from purified CD34+ bone marrow cells. Leukemia 14:874- 881. Iwata, M., A. Hirakiyama, Y. Eshima, H. Kagechika, C. Kato, and S. Y. Song. 2004. Retinoic acid imprints gut-homing specificity on T cells. Immunity 21 :527- 538. Mora, J. R., M. Iwata, B. Eksteen, S. Y. Song, T. Junt, B. Senman, K. L. Otipoby, A. Yokota, 1H. Takeuchi, P. Ricciardi-Castagnoli, K. Rajewsky, D. H. Adams, and U. H. von Andrian. 2006. Generation of gut-homing IgA-secreting B cells by intestinal dendritic cells. Science 314:1 157-1 160. Johansson-Lindbom, B., M. Svensson, O. Pabst, C. Palmqvist, G. Marquez, R. Forster, and W. W. Agace. 2005. Functional specialization of gut CD103+ dendritic cells in the regulation of tissue-selective T cell homing. J Exp Med 202:1063-1073. 132 138. 139. 140. 141. 142. 143. 144. 145. 146. 147. 148. Johansson-Lindbom, B., and W. W. Agace. 2004. Vitamin A helps gut T cells find their way in the dark. Nat Med 10: 1300-1301. Maldonado-Lopez, R., T. De Smedt, B. Pajak, C. Heirrnan, K. Thielemans, 0. Leo, J. Urbain, C. R. Maliszewski, and M. Moser. 1999. Role of CD8alpha+ and CD8alpha- dendritic cells in the induction of primary immune responses in vivo. J Leukoc Biol 66:242-246. O'Keeffe, M., H. Hochrein, D. Vremec, B. Scott, P. Hertzog, L. Tatarczuch, and K. Shortman. 2003. Dendritic cell precursor populations of mouse blood: identification of the murine homologues of human blood plasmacytoid pre-DC2 and CD1 lc+ DCl precursors. Blood 101:1453-1459. Naik, S. H., L. M. Corcoran, and L. Wu. 2005. Development of murine plasmacytoid dendritic cell subsets. Immunol Cell Biol 83:563-570. Annacker, 0., J. L. Coombes, V. Malmstrom, H. H. Uhlig, T. Boume, B. Johansson-Lindbom, W. W. Agace, C. M. Parker, and F. Powrie. 2005. Essential role for CD103 in the T cell-mediated regulation of experimental colitis. J Exp Med 202:1051—1061. Coombes, J. L., K. R. Siddiqui, C. V. Arancibia-Carcamo, J. Hall, C. M. Sun, Y. Belkaid, and F. Powrie. 2007. A functionally specialized population of mucosal CD103+ DCs induces Foxp3+ regulatory T cells via a TGF-beta and retinoic acid-dependent mechanism. J Exp Med 204:1757—1764. Gallucci, S., M. Lolkema, and P. Matzinger. 1999. Natural adjuvants: endogenous activators of dendritic cells. Nat Med 5:1249-1255. Morin, J ., A. Chimenes, C. Boitard, R. Berthier, and S. Boudaly. 2003. Granulocyte-dendritic cell unbalance in the non-obese diabetic mice. Cell Immunol 223: 1 3-25. Penna, G., S. Amuchastegui, N. Giarratana, K. C. Daniel, M. Vulcano, S. Sozzani, and L. Adorini. 2007. 1,25-Dihydroxyvitamin D3 selectively modulates tolerogenic properties in myeloid but not plasmacytoid dendritic cells. J Immunol 178:145-153. Saurer, L., K. C. McCullough, and A. Summerfield. 2007. In vitro induction of mucosa-type dendritic cells by all-trans retinoic acid. J Immunol 179:3504-3514. Carreras, E., S. Turner, V. Paharkova-Vatchkova, A. Mao, C. Dascher, and S. Kovats. 2008. Estradiol acts directly on bone marrow myeloid progenitors to differentially regulate GM-CSF or F1t3 ligand-mediated dendritic cell differentiation. J Immunol 180:727-738. 133 149. 150. 151. 152. 153. 154. 155. 156. 157. 158. 159. 160. Crowley, M., K. lnaba, M. Witmer-Pack, and R. M. Steinman. 1989. The cell surface of mouse dendritic cells: F ACS analyses of dendritic cells from different tissues including thymus. Cell Immunol 1 18:108-125. Pollard, A. M., and M. F. Lipscomb. 1990. Characterization of murine lung dendritic cells: similarities to Langerhans cells and thymic dendritic cells. J Exp Med 172:159-167. Calder, C. J ., J. Liversidge, and A. D. Dick. 2004. Murine respiratory tract dendritic cells: isolation, phenotyping and functional studies. J Immunol Methods 287:67-77. Magyarics, Z., A. Csillag, K. Pazmandi, E. Rajnavolgyi, and A. Bacsi. 2008. Identification of plasmacytoid pre-dendritic cells by one-color flow cytometry for phenotype screening. C ytometry A 73:254-258. Schlueter, A. J ., and J. K. Glasgow. 2006. Phenotypic comparison of multiple monocyte-related populations in murine peripheral blood and bone marrow. C ytometry A 69:281-290. Schlueter, A. J., S. K. Bhatia, X. Li, L. T. Tygrett, Y. Yamashita, P. de Vries, and T. J. Waldschmidt. 2001. Delineation among eight major hematopoietic subsets in murine bone marrow using a two-color flow cytometric technique. C ytometry 43:297-307. Vennaelen, K., and R. Pauwels. 2004. Accurate and simple discrimination of mouse pulmonary dendritic cell and macrOphage populations by flow cytometry: methodology and new insights. C ytometry A 61 :170-177. Walunas, T. L., D. S. Bruce, L. Dustin, D. Y. Loh, and J. A. Bluestone. 1995. Ly- 6C is a marker of memory CD8+ T cells. JImmunol 155: 1 873-1 883. Matsuzaki, J ., T. Tsuji, K. Chamoto, T. Takeshima, F. Sendo, and T. Nishimura. 2003. Successful elimination of memory-type CD8+ T cell subsets by the administration of anti-Gr-I monoclonal antibody in vivo. Cell Immunol 224:98- 105. Johansson, C., and B. L. Kelsall. 2005. Phenotype and function of intestinal dendritic cells. Semin Immunol 17:284-294. del Rio, M. L., J. I. Rodriguez-Barbosa, E. Kremmer, and R. Forster. 2007. CD103- and CD103+ bronchial lymph node dendritic cells are specialized in presenting and cross-presenting innocuous antigen to CD4+ and CD8+ T cells. J Immunol 178:6861-6866. Lukens, M. V., D. Kruijsen, F. E. Coenjaerts, J. L. Kimpen, and G. M. van Bleek. 2009. Respiratory Syncytial Virus induced activation and migration of respiratory 134 161. 162. 163. 164. 165. 166. 167. I68. 169. 170. 171. 172. dendritic cells and subsequent antigen presentation in the lung draining lymph node. J Virol. Qiu, C. H., Y. Miyake, H. Kaise, H. Kitamura, O. Ohara, and M. Tanaka. 2009. Novel subset of CD8{alpha}+ dendritic cells localized in the marginal zone is responsible for tolerance to cell-associated antigens. J Immunol 182:4127-4136. Gupta, V., and S. M. Singh. 2008. Gender dimorphism of macrophage response to GMC SF and IL-4 for differentiation into dendritic cells. Am J Reprod Immunol 60:43-54. Raju, R., K. I. Bland, and I. H. Chaudry. 2008. Estrogen: a novel therapeutic adjunct for the treatment of trauma-hemorrhage-induced immunological alterations. Mol Med 14:213-221 . Moretto, M. M., E. M. Lawlor, and I. A. Khan. 2008. Aging mice exhibit a functional defect in mucosal dendritic cell response against an intracellular pathogen. J Immunol 181:7977-7984. Stout-Delgado, H. W., X. Yang, W. E. Walker, B. M. Tesar, and D. R. Goldstein. 2008. Aging impairs IFN regulatory factor 7 up-regulation in plasmacytoid dendritic cells during TLR9 activation. J Immunol 181 :6747-6756. Asselin-Paturel, C., G. Brizard, J. J. Pin, F. Briere, and G. Trinchieri. 2003. Mouse strain differences in plasmacytoid dendritic cell frequency and function revealed by a novel monoclonal antibody. J Immunol 171:6466-6477. Roederer, M. 2001. Spectral compensation for flow cytometry: visualization artifacts, limitations, and caveats. C ytomet'ry 45:194-205. Parks, D. R., M. Roederer, and W. A. Moore. 2006. A new "Logicle" display method avoids deceptive effects of logarithmic scaling for low signals and compensated data. Cytometry A 69:541-551. Baumgarth, N., and M. Roederer. 2000. A practical approach to multicolor flow cytometry for immunophenotyping. J Immunol Methods 243:77-97. Perfetto, S. P., P. K. Chattopadhyay, and M. Roederer. 2004. Seventeen-colour flow cytometry: unravelling the immune system. Nat Rev Immunol 42648-655. Maecker, H. T., T. Frey, L. E. Nomura, and J. Trotter. 2004. Selecting fluorochrome conjugates for maximum sensitivity. Cytometry A 62:169-173. Kang, S. G., H. W. Lim, O. M. Andrisani, H. E. Broxmeyer, and C. H. Kim. 2007. Vitamin A metabolites induce gut-homing FoxP3+ regulatory T cells. J Immunol 17923724-3733. 135 173. I74. 175. 176. 177. 178. 179. 180. I81. 182. 183. 184. Stephensen, C. B., X. J iang, and T. Freytag. 2004. Vitamin A deficiency increases the in vivo development of IL-lO-positive Th2 cells and decreases development of Th1 cells in mice. J Nutr 134:2660-2666. Mucida, D., Y. Park, G. Kim, 0. Turovskaya, 1. Scott, M. Kronenberg, and H. Cheroutre. 2007. Reciprocal TH17 and regulatory T cell differentiation mediated by retinoic acid. Science 317:256-260. Steinman, R. M., M. Pack, and K. Inaba. 1997. Dendritic cells in the T-cell areas of lymphoid organs. Immunol Rev 156:25-37. Darrnanin, S., J. Chen, S. Zhao, H. Cui, R. Shirkoohi, N. Kubo, Y. Kuge, N. Tamaki, K. Nakagawa, J. Hamada, T. Moriuchi, and M. Kobayashi. 2007. All- trans retinoic acid enhances murine dendritic cell migration to draining lymph nodes via the balance of matrix metalloproteinases and their inhibitors. J Immunol 179:4616-4625. Lackey, D. E., S. L. Ashley, A. L. Davis, and K. A. Hoag. 2008. Retinoic acid decreases adherence of murine myeloid dendritic cells and increases production of matrix metalloproteinase-9. J Nutr 138:1512-1519. Erhardt, J. G., H. K. Biesalski, L. C. Malaba, and N. E. Craft. 2003. Improved method for measurement of retinol and didehydroretinol in the modified relative dose response test to detect vitamin A deficiency. Clin Chem 49:338-339. Duriancik, D. M., and K. A. Hoag. 2009. The identification and enumeration of dendritic cell populations from individual mouse spleen and Peyer's patches using flow cytometric analysis. C ytometry A 75:951-959. Chandra, R. K. 1983. Numerical and functional deficiency in T helper cells in protein energy malnutrition. Clin Exp Immunol 51:126-132. Fraker, P. J ., L. E. King, T. Laakko, and T. L. Vollmer. 2000. The dynamic link between the integrity of the immune system and zinc status. J Nutr 130: 1 3998- 14068. King, L. E., F. Osati-Ashtiani, and P. J. Fraker. 2002. Apoptosis plays a distinct role in the loss of precursor lymphocytes during zinc deficiency in mice. J Nutr 132:974-979. Semba, R. D., Muhilal, B. J. Ward, D. E. Griffin, A. L. Scott, G. Natadisastra, K. P. West, Jr., and A. Sommer. 1993. Abnormal T-cell subset proportions in vitamin-A-deficient children. Lancet 341 :5-8. Kinoshita, M., A. M. Pasatiempo, C. E. Taylor, and A. C. Ross. 1991. Immunological memory to tetanus toxoid is established and maintained in the vitamin A-depleted rat. F aseb J 5:2473-2481. 136 185. 186. 187. 188. 189. Winau, F ., G. Hegasy, R. Weiskirchen, S. Weber, C. Cassan, P. A. Sieling, R. L. Modlin, R. S. Liblau, A. M. Gressner, and S. H. Kaufmann. 2007. Ito cells are liver—resident antigen-presenting cells for activating T cell responses. Immunity 26:117-129. Sparrnann, G., A. Glass, P. Brock, R. Jaster, D. Koczan, H. J. Thiesen, S. Liebe, and J. Emmrich. 2005. Inhibition of lymphocyte apoptosis by pancreatic stellate cells: impact of interleukin-15. Am J Physiol Gastrointest Liver Physiol 289:G842-851. Jonuleit, H., K. Wiedemann, G. Muller, J. Degwert, U. Hoppe, J. KnOp, and A. H. Enk. 1997. Induction of IL-15 messenger RNA and protein in human blood- derived dendritic cells: a role for IL-15 in attraction of T cells. J Immunol 158:2610-2615. DaWSOn, H. D., G. Collins, R. Pyle, M. Key, A. Weeraratna, V. Deep-Dixit, C. N. Nadal, and D. D. Taub. 2006. Direct and indirect effects of retinoic acid on human Th2 cytokine and chemokine expression by human T lymphocytes. BMC Immunol 7:27. Wendland, M., N. Czeloth, N. Mach, B. Malissen, E. Kremmer, O. Pabst, and R. Forster. 2007. CCR9 is a homing receptor for plasmacytoid dendritic cells to the small intestine. Proc Natl Acad Sci U S A 104:6347-6352. 137 Mlclmlelllllllllilllillill 3 1293 03063 V LIBRARIES 4962