mmmmg. .37.! .31 BI. .50 .VI... 9 . I! 5.! ‘4‘ CI flit ¥ .. SLE .nnu.uunumny.;.\mnz 1.4 u... . 3...... '1 xvi. itgis 2 1 3 1‘ {I ’3 :E ‘3 fiffi'fib’fih (,1 (AI I‘J ,3 LIBRARY Michigan State Ul liversity This is to certify that the dissertation entitled INFLAMMATORY SIGNALING IN MACROPHAGES: REGULATION BY G-PROTEIN COUPLED RECEPTOR KlNASE-2 AND 5 presented by SONIKA PATIAL has been accepted towards fulfillment of the requirements for the Ph.D. degree in Cell and Molecular Biology MW M 'or Professor’s Signature 0?.0‘21f‘a20/O Date MSU is an Affirmative Action/Equal Opportunity Employer I u-l' '4... d. ‘I I 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 KzlProj/AccauPres/CIRCIDateDue.indd INFLAMMATORY SIGNALING IN MACROPHAGES: REGULATION BY G- PROTEIN COUPLED RECEPTOR KlNASE-2 AND 5 By Sonika Patial A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Cell and Molecular Biology 2010 ABSTRACT INFLAMMATORY SIGNALING IN MACROPHAGES: REGULATION BY G- PROTEIN COUPLED RECEPTOR KINASE-Z AND 5 By Sonika Patial G-protein coupled receptor kinases (GRKs) are serine-threonine protein kinases which phosphorylate agonist bound G-protein coupled receptors (GPCRs) leading to their desensitization. Although originally discovered with regard to GPCR desensitization, recent studies have shown that GRKs play much wider roles than previously appreciated. In this regard, studies have shown that GRKs can phosphorylate non GPCR receptor as well as non receptor substrates. There is an ample amount of evidence in the literature showing that the expression and activity of GRKs is altered in several inflammatory disease conditions. For instance, the activity and expression levels of GRK2 were found to be significantly decreased in peripheral blood mononuclear cells (PBMC) of patients with rheumatoid arthritis whereas the expression levels of GRK2 and GRKS were found to be enhanced in the neutrophils of sepsis patients. Moreover in our previous studies we found that stimulation of primary peritoneal macrophages with LPS under in vitro conditions causes an increase in the expression of GRK2. These studies point to a crucial role of GRKs in inflammatory signaling, however, the physiological importance of changes in the expression levels of GRKs is not well established. To enhance our understanding of the role of GRK2 and 5 in inflammatory signaling, we first investigated the mechanism by which GRK2 and 5 regulate TNFa-induced inflammatory signaling in mouse macrophage cell line. Our results in this study demonstrated that both GRK2 and 5 positively regulate TNFa-induced NFKB signaling. Knockdown of GRK2 and 5 inhibited TNFa-induced NF KB signaling whereas overexpression of GRK2 and 5 substantially enhanced TNFa- induced NFKB activity. GRK2 and 5 were found to interact with and phosphorylate IKBa, an inhibitor of NF KB and this was found to be the biochemical mechanism of regulation of NFKB signaling pathway by GRK2 and 5. To further elucidate the role of GRKs in inflammation under in vivo conditions, we utilized mice with homozygous GRKS gene deletion. Primary peritoneal macrophages from GRK5"' mice stimulated with LPS showed an inhibition of NFKB activity as compared to cells from GRKSH+ mice. Secretion of several LPS-induced inflammatory cytokines was found to be reduced in cell culture supernatants of GRK5"' mice. Plasma levels of cytokines and chemokines were also found to be reduced which was associated +/+ with reduced liver injury in GRK5'/' mice compared to GRKS mice suggesting that GRKS positively regulates LPS-induced inflammatory signaling in vivo by modulating transcription factor NFKB. Since homozygous gene deletion of GRK2 is lethal in mice, we utilized Cre-loxP system to achieve a cell specific deletion of GRK2 whereby GRK2 was specifically deleted in the cells of myeloid lineage. LPS injection into these mice caused an increased expression of cytokines / chemokines in the plasma as well as in the primary peritoneal macrophage cell culture supematants. GRK2 deficient mice also exhibited an increased lung and liver injury. Mechanistically, IKKB-NFKBI p105 f1“ PL2- MEK-ERK pathway was found to be negatively regulated by GRK2 which resulted in an increased expression of cytokines / chemokines in GRK2 deficient mice. Taken together, these results show that both GRK2 and 5 play a crucial role in LPS-induced inflammatory signaling under in vivo conditions by regulating NFKB and ERK signaling pathways. ACKNOWLEDGMENTS I would like to express my profound gratitude to my advisor Dr. Nara Parameswaran who gave me the opportunity to be his student. I want to thank him for his generous time, patience, sincere guidance, continuous encouragement and support. His precious scientific advice and great attitude towards science compelled me to adopt those elite qualities in my deve10ping scientific career which I will take with me through my entire career. I feel extremely fortunate to have Dr. Parameswaran as my major advisor for whom no words of praise are sufficient. I also deeply acknowledge my guidance committee members; Dr. Karl Olson Dr. Kathleen A Gallo, Dr. Andrea Amalfitano and Dr. William Spielman for their great help, insightful comments, constructive discussions and continual support. I am indebted to them for their valuable suggestions and continued inputs to improve my research progress as well as my scientific career. I am also highly thankful to Dr. Patricia Senagore for her help in pathology studies. I am thankful to the Cell and Molecular Biology Program, Dr. Susan Conrad (Director, Cell and Molecular Biology Program) and Dr. Barbara Sears (Director, Genetics Program) for accepting me into their programs and giving me this great opportunity to enhance my scientific career. I sincerely thank Becky Manse], Jeannine Lee and Christine VanDeuren for all their help and making every administrative work look so simple starting from the day I submitted my application to the graduate school. I am deeply obliged to Shipra, Katie and Sita for their help in my research work. I would like to thank all the lab members from Dr. Amalfitano’s laboratory, especially Daniel Appledom, Sergey Seregin and Sarah Godbehere for their endless help during my research work. My sincere thanks are extended to all current and former colleagues in Dr. Parameswaran’s laboratory, especially Katie Porter, Shipra Shahi, Megan Hull, Wen Qin, Nandita, Taehyung, Sita Ram and Babu Gonipeta for all their kind help, cooperation, and friendly atmosphere that they provided me. I also want to thank all my friends and colleagues in Cell and Molecular Biology Program, Physiology department and Genetics Program. I am greatlyappreciative of my friends here at MSU specially Stancy, Madalina, Neli, Navneet, Hey-jin, Michelle, Vishal, Dorothy, Krista, Scott, Nitin, Chris, Fei, Amal, Miha, Sunetra, Aparajita, Ram and Anita for their kind help and support during all these years. I greatly appreciate Josselyn, Barb, Carolyn, Richard and Kim for their support and help throughout my stay in the physiology department and making me feel at home. I greatly appreciate ULAR staff and Human Histopathology Division specially Amy and Kathy for their help in my research work. I am thankful with all my heart to my parents for their encouragement, love and support which motivated me to accomplish my educational goals. Finally, my special thanks go to my dear husband, Yogesh who has been a source of motivation during all those tough moments. It is a very special time of my life as we were blessed with our baby boy, Krish on November IS, 2009. TABLE OF CONTENTS LIST OF TABLES ............................................................................... ix LIST OF FIGURES .............................................................................. x KEY TO ABBREVIATIONS ................................................................. xiii CHAPTER 1: ...................................................................................... 1 Introduction ............................................................................. 1 CHAPTER 2: ..................................................................................... 3 Literature Review ..................................................................... 3 2.1: G protein coupled receptor kinases (GRKs) ................................ 3 2.1.1: Historical perspectives of GRK discovery .................................. 3 2.1.2: GRK mediated desensitization of GPCRs ................................... 4 2.1.3: Structure and distribution of GRKs .......................................... 5 2.1.4: Regulation of GRKs by other proteins ..................................... 12 2.1.4.1 Regulation of GRKS by Caveolin, Clathrin and a-actinin ............ 12 2.1.4.2 Regulation of GRKs by Calcium binding proteins ..................... 13 2.1.4.3 Regulation of GRKs through phosphorylation by other kinases.....14 2.1.4.4 Regulation of GRKs by GM subunits and phospholipids ............ 16 2.1.4.5 Regulation of GRKs by interaction with agonist activated GPCRs.17 2.1.5: Other novel interactions of GRKs and their physiological effects......18 2.2 Physiological and Pathological roles of GRKs ............................. 22 2.2.1: Physiological roles of GRKs as determined by knockout mice ......... 23 2.2.2: Pathological roles of GRKs ................................................... 25 2.3: GRKs and inflammation ....................................................... 29 2.3.1: GRKs expression and inflammation ......................................... 29 2.3.2: GRKs deletion and inflammation ............................................ 30 2.3.3: GRKs regulation of chemokine signaling and inflammation ............ 31 2.4: LPS and TNFa induced inflammation ...................................... 31 2.4.1 LPS induced inflammation ................................................... 31 2.4.1.1: Toll like receptor 4 (TLR4) signaling .................................... 34 2.4.2 TNFa-a potent pro-inflammatory cytokine .................................. 36 vi 2.5: N F-KB transcription factor in inflammation ............................... 41 2.6: MAPIQ in inflammation ....................................................... 48 2.6.1: ERK1/2 kinase ................................................................. 48 2.6.2: P38 MAPK ..................................................................... 49 2.6.3: JNK kinase ..................................................................... 51 2.7: Septic shock ...................................................................... 51 2.7.1 Animal models of sepsis ...................................................... 52 2.7.2 Different animal species as models for sepsis ............................. 56 2.7.3 Neutrophils and Monocyte / macrophages in sepsis ...................... 56 2.7.4 Coagulation defect in sepsis .................................................. 57 2.7.5 Multiple organ failure ........................................................ 58 Hypothesis and Specific Aims ....................................................... 60 References .............................................................................. 62 CHAPTER 3: G—protein coupled receptor kinases mediate TNFa—induced NFKB signaling via direct interaction with and phosphorylation of IKBa .................................... 80 Abstract ................................................................................ 81 Introduction ............................................................................ 82 Material and Methods ................................................................ 85 Results .................................................................................. 91 Discussion ............................................................................ 128 References ............................................................................. 1 3 3 CHAPTER 4: G-protein coupled receptor kinase 5 (GRKS) mediates Toll-like receptor-4 -induced NFKB pathway in macrophages and is necessary for the production of inflammatory cytokines and chemokines in viva ........................................ 139 Abstract ............................................................................... 140 Introduction ........................................................................... 141 Material and Methods ............................................................... 144 Results ................................................................................. 147 Discussion ............................................................................ 1 77 References ............................................................................. 1 82 CHAPTERS: vii Myeloid specific G-protein coupled receptor kinase 2 (GRK2) is a negative regulator of NFchl p105-ERK pathway and limits endotoxemic shock in mice..186 Abstract ................................................................................ 187 Introduction ............................................................................ 1 88 Material and Methods ............................................................... 191 Results ................................................................................. 195 Discussion ............................................................................ 240 References ............................................................................. 248 CHAPTER 6: Summary and Conclusions ................................................................... 252 6.1 Specific aims and results of the study ..................................... 252 6.2 Limitations of the study ....................................................... 254 6.3 Positive outcomes of the study ............................................. 255 6.4 Future directions ............................................................... 257 viii Table 2.1 Table 2.2 Table 2.3 Table 2.4 LIST OF TABLES Classification of G-Protein coupled receptor kinases (GRKs)............7 Phenotypic characteristics of GRK Knockout/Transgenic mice ....... 27 Involvement of GRKs in diseases ............................................ 32 Animal models of sepsis and their advantages verses disadvantages .................................................................. 54 Figure 2.1 Figure 2.2 Figure 2.3 Figure 2.4 Figure 3.1 Figure 3.2 Figure 3.3 Figure 3.4 Figure 3.5 Figure 3.6 Figure 3.7 Figure 3.8 Figure 3.9 Figure 3.10 Figure 3.11 LIST OF FIGURES Images in this dissertation are presented in color Comparison of Linear Structure of GRK5 ................................. 9 LPS activates NF KB and MAPK pathways ............................... 37 TNFa stimulates NFKB and MAPK pathways ............................ 39 Mammalian NFKB and IKE family members.................................43 GRK2 knockdown in macrophages inhibits TNFa-induced IKBO. phosphorylation and degradation ..................................... 93 Effect of different GRK2 siRNA oligos on TNFa-induced IKBa phosphorylation ....................................... 96 GRK2 overexpression enhances TNFa-induced Icha phosphorylation in macrophages ............................................ 98 GRK5 knockdown inhibits TNFa-induced IKBO. phosphorylation and degradation in macrophages ...................... 101 Effect of a different GRK5 siRNA oligo on TNFa-induced IKBa phosphorylation ..................................... 104 Role of GRKs is specific for TNFa- NFKB pathway .................................................................. 107 Expression of macrophage inflammatory protein-113 (MIPI B), an NFKB-regulated gene, is inhibited by GRK2 or GRK5 knockdown in macrophages ................................................ 109 Kinase activity of GRK2 and GRK5 is essential for mediating TNFa-induced NFKB transcriptional activation ............ 111 GRK2 interacts with the N-terminus of IKBO. .......................... 115 Direct interaction between GRK5 and IKBO. ............................ 117 IKBa is a substrate for GRK2 and GRK5 in vitro ....................... 120 Figure 3.12 Figure 4.1 Figure 4.2 Figure 4.3 Figure 4.4 Figure 4.5 Figure 5.1 Figure 5.2 Figure 5.3 Figure 5.4 Figure 5.5 Figure 5.6 Figure 5.7 Figure 5.8 Figure 5.9 IKKB knockdown does not inhibit TNFa-induced IicBa phosphorylation but inhibits LPS-induced phosphorylation ........... 125 GRK5 null peritoneal macrophages show reduced TLR4-induced NFKB activation .............................................................. 149 GRK5 has no effect on TLR4—induced MAPK activation ............. 154 LPS induction of cytokines and chemokines in peritoneal macrophages from GRK5“+ and GRK5"' mice .......................................... 159 Profile of plasma cytokines after LPS challenge in GRK5”+ and GRK5"’ mice .................................................................. 167 GRK5"' mice show reduced LPS-induced liver injury.................. 1 74 mye . . . A . Generation and characterization of GRK2 mice .................. 197 Complete blood count of cells from GRK2Amye verses GRK2fl/fl mice ........................................................................... 199 LPS injection causes an enhanced secretion of pro and anti- ye inflammatory cytokines in the plasma of GRK2Am mice .......... 204 Amye . . . . . GRK2 mice show enhanced tissue injury and mortality as fl/fl . compared to GRK2 mice ............................................... 211 . Am e , Peritoneal macrophages fi'om GRK2 y mice secrete enhanced amounts of pro-inflammatory cytokines and chemokines in response to LPS stimulation .............................................................. 216 . A . Neutrophils from GRK2 mye mice secrete enhanced amounts of pro- inflammatory cytokines and chemokines in response to LPS stimulation .................................................................... 219 GRK2 deficiency causes an enhanced LPS stimulated ERK1/2 activation in peritoneal macropahges ..................................... 224 GRK2 deficiency does not affect LPS stimulation of Akt and GSK3B activation in peritoneal macrophages ..................................... 227 The existence of IKKB-NFKBI p105/Tp12-MEK-ERK pathway in peritoneal macrophages confirmed by using an IKKB inhibitor...... 230 xi Figure 5.10 Figure 5.11 GRK2 deficiency causes an enhanced LPS stimulated NFKBI p105 activation in peritoneal macrophages ..................................... 233 Pharmacological inhibition of ERK and IKKB causes a significant inhibition of LPS-induced secretion of cytokines and chemokines in A . peritoneal macrophages from GRK2 mye mice ....................... 236 xii ANOVA ATCC ATP cDNA CK CMV CRE DTT EDTA EMSA GPCR GRK GRK2 GRK5 GST HA HEK HEPES HRP IAPI KEY TO ABBREVIATIONS Analysis of Variance American Type Culture Collection Adenosine triphosphate Complimentary DNA Casein Kinase Cytomegalovirus Cyclization recombination Dithiothreitol Ethylenediaminetetraacetic acid Electrophoretic mobility shift assay G-Protein coupled receptor G-Protein coupled receptor kinase G-Protein coupled receptor kinase-2 G-Protein coupled receptor kinase-5 Glutathione S-transferase Hemaglutinin Human Embryonic Kidney N-2-Hydroxyethylpiperazine-N'-2-Ethanesulfonic Acid Horseradish peroxidase Inhibitor of apoptosis 1 xiii IKKB IL IPTG IKBa KCI LB LoxP LPS MgC12 ml mM mRNA NaCl NFKB PBS RIP 1 RT RT-PCR SDS-PAGE IKB kinase B Interleukin Isopropyl B-D-l-thiogalactopyranoside Inhibitor of NFKB Potassium Chloride Luria Bertani Locus of crossover in P1 Lipopolys‘accharide Magnesium Chloride Milliliter Millimolar Messenger RNA Sodium Chloride Nuclear factor-kappa B Nanomolar Phosphate Buffered Saline Presence of active hydrogen Receptor interacting protein 1 Reverse Transcriptase Reverse transcriptase-polymerase chain reaction Sodium dodecyl sulfate polyacrylamide gel electrophoresis xiv SEM Standard error of mean TAD Transactivation domain TBS-T Tris—Buffered Saline-Tween 20 TNF R TNF receptor TNFa Tumor necrosis factor-alpha TRADD Tumor necrosis factor receptor 1 associated death domain protein TRAF TNF receptor associated factor UV Ultraviolet pg Micro gram pl Microliter um . Micrometer XV CHAPTER 1 INTRODUCTION G protein coupled receptor kinases (GRKs) are serine threonine kinases which play an important role in the desensitization of G protein coupled receptors (GPCRs). Agonist bound GPCRs are a target for phosphorylation by GRK5, thus leading to their rapid homologous desensitization. Although originally discovered with regard to this classical role of GPCR desensitization, recent studies have shown that GRK5 can phosphorylate non-GPCR receptor as well as non-receptor substrates. Moreover, GRKs have also been shown to be involved in other phosphorylation-independent protein-protein interactions. In this regard, GRKs have been shown to regulate Toll ' like receptor 4 (TLR4) induced inflammatory signaling in macrophages [1]. Thus, it was hypothesized that GRK5 are important regulators of inflammatory signaling in macrophages and therefore GRK5 might play essential roles in the regulation of inflammatory diseases. This thesis research investigates the role of GRKs in inflammatory signaling mediated by TNF receptors in macrophages under in vitro conditions as well as the role of GRKs in TLR4-induced inflammatory signaling under in vivo conditions in a mouse model of sepsis. This brief introduction provides an overview of the hypothesis tested in this thesis with a glance on the focus of subsequent chapters. Chapter two provides a comprehensive literature review which provides an in-depth discussion of the scientific literature surrounding the focus of this thesis project, including the classical and novel roles of GRK5 as well as the involvement of GRK5 in inflammation and immune response. Chapters three through five are organized based on the specific aims of this thesis research project, each consisting of an abstract, introduction, materials and methods, results and discussion relevant to the individual aim. Chapter three focuses on the role of GRK5, in particular GRK2 and GRK5, in TNFa induced inflammatory signaling particularly in mouse macrophages under in vitro conditions. Chapter four investigates the role of GRK5 at the in vivo level in a mouse model of LPS induced sepsis using a homozygous GRK5 gene deletion. Chapter five tests the role of GRK2 in inflammatory signaling at the in vivo level whereby the focus was to generate a myeloid cell specific genetic deletion of GRK2 and investigating the role of this conditional deletion in a mouse model of LPS induced sepsis. Chapter six highlights the results obtained in this thesis project with a discussion on how these findings contribute to our current understanding of the novel roles of GRK5 as well as the important role of GRK5 in LPS mediated sepsis. Also, discussed are some of the limitations of this project as well as future directions. CHAPTER 2 LITERATURE REVIEW 2.1: G protein coupled receptor kinases (GRK5) G-protein coupled receptor kinases (GRK5) are serine/threonine kinases which were originally discovered with regard to G-protein coupled receptor (GPCR) desensitization [2]. They are the key modulators of GPCR signaling and play an important role in fine tuning the GPCR signaling mechanism preventing the overstimulation of GPCRs even in the presence of continuous agonist stimulation. 2.1.]: Historical perspectives of GRK discovery Studies of mechanisms involved in the homologous desensitization of [32- adrenergic receptor (BZAR) and rhodopsin (the prototypic light receptor) led to the discovery of GRK5. In particular, rhodopsin played an important role in our current understanding on the desensitization mechanism of GPCRs by GRKs due to its availability in greater quantities. Rhodopsin undergoes light dependent phosphorylation, first discovered in vivo in the 1970’s [3, 4]. Subsequently, it was shown that the kinetics of rhodopsin phosphorylation correlate with the quenching of cGMP phosphodiesterase activity, suggesting that rhodopsin phosphorylation has a role in its desensitization [5]. Rhodopsin kinase (GRKl) was then purified and shown to phosphorylate the receptor at multiple serine and threonine residues. The occurrence of agonist-induced phosphorylation and desensitization of BZAR even in the absence of protein kinase A (in kin' S49 lymphoma cells) and alpha subunit of Gs (in cyc' cells) suggested the presence of another kinase, subsequently identified as B-adrenergic receptor kinase (BARK) [6, 7]. In the mid 1980’s, BARK was also discovered as a functional homologue to rhodopsin kinase and this further suggested the existence of a multigene family of protein kinases which phosphorylate agonist bound GPCRs [8]. Subsequently, crude BARK preparations were made and were shown to mediate agonist dependent phosphorylation and uncoupling of the receptor in vitro. A cDNA encoding BARK (or GRK2) was subsequently sequenced and cloned, which expressed an enzyme that preferentially phosphorylates the agonist- bound B2AR [9]. Pure preparations of BARK however did not lead to a significant loss of Gs coupling. This suggested that an essential cofactor needed for BARK mediated desensitization was lost during the purification process. This essential cofactor was subsequently discovered as an additional protein called arrestin. Four distinct members of arrestins are now known. Two out of these are restricted to phototransduction pathways (visual arrestinl and visual arrestin 2). The other two i.e beta arrestin 1 (arrestin 2) and beta arrestin 2 (arrestin 3) are expressed ubiquitously and regulate the internalization of many GPCRs as well as also play an important role in cellular signaling [10]. 2.1.2: GRK mediated desensitization of GPCRs GRKs cause homologous desensitization of the receptors whereby only the agonist activated receptors are phosphorylated and desensitized [2],[11]. Agonist- induced desensitization of a GPCR is a multistep process. GRKs first phosphorylate agonist bound GPCRs on serine and threonine residues located in the carboxy-terminal tail region and/or the third cytoplasmic loop. This process enhances the affinity of agonist bound-GPCR for binding to arrestins which then sterically inhibit the interaction of the receptors with the G-proteins [12] leading to rapid homologous desensitization. Finally, the GRK-arrestin system induces the internalization of inactivated receptors to endosomal compartments via endocytosis. Endocytosis of receptors has a fundamental role in receptor biology although this process is not necessary for homologous desensitization [13]. Intemalization of receptors can have three consequences viz. (1) Dephosphorylation of the receptor, leading to resensitization and recycling back to the cell surface (2) Targeting of the receptor to lysosomes leading to degradation (3) or activation of other signaling pathways. Furthermore, endocytosis can occur via clathrin coated pits, caveolea or other uncoated vesicles.[14, 15]. For clathrin dependent internalization, phosphorylation of receptors by GRKS and subsequent binding by arrestins appears to be essential [12]. Moreover, arrestin and clathrin dependent endoycytosis is used by a majority of GPCRs. 2.1.3: Structure and distribution of GRK5 The GRK family consists of seven different genes and based on sequence and functional similarities they are divided into three subfamilies [16]. The rhodopsin kinase subfamily comprises GRKI (rhodopsin kinase) and GRK7 (cone opsin kinase) that are found exclusively in retinal cells. The GRK2 subfamily comprises pleckstrin homology domain containing GRK2 (B-ARKl) and GRK3 (B-ARKZ). Their membrane recruitment depends on interaction with the GBy subunits of G proteins and phosphatidyl-inositol 4,5—biphosphate. The GRK4 subfamily comprises GRK4, GRK5 and GRK6 which are predominantly localized at the membrane either due to palmitoylation on C-terminal cysteine residues (GRK4/6) or interaction between a positively charged domain at the C-terminus with the negatively charged membrane phospholipids (GRK5) [17]. GRK4 is predominantly found in testes [18] and to a lesser extent also in brain and kidney [19] whereas GRK2, GRK3, GRK5 and GRK6 are widely expressed (Table 2.1). All GRK5 have a similar structural organization possessing an N-terminal domain, a central catalytic domain (with homology to other serine-threonine kinases) and a C-terminal domain. The N-terminal domain of 183-188 amino acids includes a region of homology to regulators of G-protein signaling (RGS) proteins that are known to act as GTPase activating proteins for many Ga subunits. The carboxyl terminal domain is of variable length and contains key determinants important for localization and translocation to the membrane either by post translational modifications or by sites of interaction with lipids or membrane proteins. In this regard, GRKl and GRK7 are isoprenylated, GRK4 and GRK6 are palmitoylated at one or more cysteine residues clustered within the last 15-20 amino acids of the C- terminus, which is responsible for their membrane localization whereas GRK5 is localized in the membrane by virtue of a phosphatidyl inositol 4,5 biphosphate binding Table 2.1: Classification of G protein coupled receptor kinases (GRK5) GRK5 have been subdivided into three subfamilies based on the sequence and functional similarities. 1. Rhodopsin kinase subfamily which comprises GRKI (Rhodopsin kinase ) and GRK7 (cone opsin kinase). 2. GRK2 subfamily which comprises GRK2 (B-ARKl) and GRK3 (B-ARKZ). 3. GRK4 subfamily which comprises GRK4, GRK5 and GRK6. Rhodopsin kinase subfamily GRKI GRK7 GRK2 (B-ARKI) BARK subfamily GRK3 (B-ARKZ) GRK4 GRK4 subfamily GRK5 GRK6 Figure 2.1: Comparison of Linear structure of GRKs The seven isoforms of GRKs share a number of structural and functional similarities. The amino terminal domain (183-188 amino acids) includes a region of homology to regulators of G protein signaling (RGS) proteins. The central catalytic domain has a homology to other serine threonine kinases. The carboxyl terminal domain is of variable length and contains several key determinants for the localization and translocation of kinases to the membrane. This occurs either through post translational modifications or the presence of sites of interaction with lipids or membrane proteins. As shown in the figure, GRKI and GRK7 undergo isoprenylation; GRK4 and GRK6 undergo palmitoylation: GRK5 has positively charged amino acid clusters at the C- terminus through which it binds to membrane phospholipids: GRK2 and GRK3 bear an extended C-terminus known as Pleckstrin homology domain (PH domain). This domain modulates the membrane targeting of GRK2 and GRK3. PL, phospholipids; auto (:h), stimulatory or inhibitory autophosphorylation sites. >=E£n=w 335 3.2.3 50 3:23 .E 9.10 >=E£n=m 9.55.. >8 9:25 .E v3.4a Eanu In. $155.0 ages...» 32: l 5on 0 i536 . u 5. 52:2. 35:55.0 Essen 2583 52:29 mum 10 domain at the N-terminus and a polybasic region (amino acids 547—560) close to the C-terminus [20]. GRK2 and GRK3 hear an extended C-terminus which is involved in the modulation of the kinase targeting to the membrane. GRK2 and GRK3 are cytoplasmic proteins that translocate to the membrane upon receptor stimulation. Their C-terminus harbors a pleckstrin homology (PH) domain (residues 561-655) that partially overlaps with the GBy-binding region. It has been shown that free GBy sub- units can bind to GRK2 with a high affinity. Furthermore it has been shown that GBy is required for association of GRK2 to lipid vesicles in reconstituted systems [21]. Hence, it is believed that the interaction of GBy with GRK2 and GRK3 targets them to membranes at the site of GPCR activation. Binding of GBy also enhances the activity of GRK2 by causing allosteric activation of this kinase. The PH domain of GRK2 and GRK3 also interacts with phosphatidylinositol 4,5-biphosphate (PIP2) and other acidic phospholipids such as phosphatidyl serine (PS) which regulate its kinase activity [22]. Thus, GBy and lipids contribute synergistically to membrane localization and activation of GRK2 (Figure 2.1). The GRK4-6 family of GRKs has been shown to contain a nuclear localization sequence (NLS). It was found that the nuclear localization of GRK5 is regulated by GPCRs. Binding of an agonist onto a GPCR leads to an elevation of intracellular calcium levels and activates a calcium sensor protein, calmodulin. Furthermore, GRK5 was found to bind to DNA under in vitro conditions [23]. More recently, it was shown that GRK5 can act as a histone deacetylase (HDAC) kinase independent of its actions on GPCRs [24]. ll 2.1.4: Regulation of GRKs by other proteins 2.1.4.1 Regulation of GRK3 by Caveolin, Clathrin and a—actinin Caveolins control the basal kinase activity of GRKs: Caveolin is a 22-24 kDa integral membrane protein and is a prominent structural component of caveolae. Caveolae are specific cholesterol and glycosphingolipid-enriched plasma membrane structures in cells. A 20 a region within the N-terminal domain of caveolin (known as scaffolding domain) has been found to mediate the association of caveolin with other proteins. Caveolins serve as scaffolding proteins for some GPCRs as well as certain mitogen activated protein kinases (MAPKs) and G proteins and help compartrnentalize signaling pathways. Recent studies have shown that GRK2 is present in caveolin rich fractions of cellular membranes. GRK2 contains caveolin binding motifs in the PH domain (residues 567-584) as well as in the N-terminal domain (residues 63-71) while GRK3 and GRK5 can bind to caveolin only through N- terminal motifs. GRK2 binding to caveolin 1 or 3 inhibits GRK2 mediated phosphorylation suggesting caveolins can control the basal kinase activity of GRK2. In addition, it is possible that GRK-caveolin interactions may facilitate GRK5 interactions with other signaling molecules [25]. Clathrin promotes arrestin independent internalization: Clathrin is a protein present in the internalized vesicles containing GPCRs. GRK2 can also interact with clathrin protein via a clathrin box located in the carboxyl terminal region of GRK2 [26]. B-arrestins bind to phosphorylated GPCRs and promote the internalization of the receptors by interacting with clathrin. Certain GPCRs, however, are only 12 slightly internalized due to their low affinity binding to B-arrestins. GRK2 bound to such receptors can facilitate GPCR internalization due to its ability to bind clathrin. This internalization is however, B-arrestin independent but depends on the presence of dynamin. [27]. Alpha-actinin inhibits GRKs: a-actinin is a protein that falls under the spectrin superfamily of actin crosslinking proteins. It is found most abundantly in muscle cells. It binds and crosslinks actin as well as lipid, creating a submembraneous meshwork consisting of crosslinked actin. a-actinin also binds various ion channels, cell adhesion molecules as well as transmembrane receptors, thereby incorporating them into the actin network creating signaling domains. It has been found that all GRK5 can interact with a-actinin. a-actinins can completely inhibit GRK3, or modulate their activity, as well as substrate specificity [28]. 2.1.4.2 Regulation of GRK5 by Calcium binding proteins Recoverin inhibits GRK]: Recoverin is a protein present in vertebrate photoreceptors, certain retinal cone bipolar cells and pineal glands, exhibiting a distribution quite similar to GRK]. Recoverin binds directly to GRKI and inhibits it in 2+ . . . . . . a Ca -dependent fashion. Calcrum levels are very high during dark conditions and fall substantially under light conditions in the vertebrate photoreceptor systems. Hence, GRK] would be complexed to recoverin and inactive when rhodopsin is in its inactive state under dark conditions. However, following rhodopsin activation, a drop in intracellular calcium levels causes the release of GRK] from recoverin. GRKI is now 13 disinhibited and can facilitate rapid phosphorylation and desensitization of rhodopsin [29]. Calmodulin inhibits GRKs: Calmodulin is a universal mediator of calcium signaling, which has been shown to inhibit the activity of GRK2, 3, 4, 5, and 6 albeit with different potencies [30]. Calmodulin binds directly to both GRK2 and GRK5, with different affinities, inhibiting the interaction of these enzymes with their agonist occupied receptors. GRK5 is highly sensitive to the presence of calcium bound calmodulin (IC50 ~ 50nM), whereas GRK2 is affected only at higher concentrations (IC50 ~ ZuM) [31]. Thus GRK2 and GRK5 mediated desensitization of GPCRs would + be predicted to be inhibited in the presence of Ca2 /calmodulin. GRK2 has a relatively low affinity for calmodulin suggesting that inhibition of GRK2 would occur only at sites highly enriched in calmodulin. However, the relatively high affinity of GRK5 for calmodulin suggests that this regulatory mechanism might be present at multiple locations. 2.1.4.3 Regulation of GRK5 through phosphorylation by other kinases Protein Kinase C (PKC) activates GRK2 but inhibits GRK5: GRK5 activity, the protein stability as well as their ability to interact with other proteins is highly regulated by their own phosphorylation by other protein kinases. In this regard protein Kinase C (PKC) has been shown to phosphorylate both GRK2 and GRK5 in intact cells as well as under in vitro conditions. PKC phosphorylation of GRK2 enhances the translocation and interaction of GRK2 with the plasma membrane causing an increased activity towards receptor substrates. It does not, however, affect 14 the catalytic activity of GRK2 since GRK2 cannot phosphorylate soluble peptides under similar conditions. PKC phosphorylates GRK2 at serine 29 within the calmodulin binding motif of GRK2 [32]. It has been suggested that the inhibitory effect produced by calmodulin binding to GRK2 is abolished after phosphorylation by PKC, allowing the binding of GRK2 to the receptor substrate. In contrast to this, phosphorylation of GRK5 by PKC leads to the inhibition of its activity towards both receptors and soluble substrates suggesting a direct effect on the catalytic activity of GRK5. There are two major sites of phosphorylation by PKC in the C-terminal 26 amino acids of GRK5. The inhibitory autophosphorylation sites also reside in this region which are targeted in the presence of calmodulin. Thus Ca/Calmodulin and PKC both act as inhibitors by targeting identical inhibitory sites on GRK5. [20]. Protein kinase A (PKA) activates GRK2: Protein kinase A (PKA) is another kinase that can phosphorylate GRK2 at serine 685. This site is located near the GBy binding domain of GRK2. PKA phosphorylation of GRK2 at this site enhances the binding of GRK2 with the GBy subunits, facilitating the membrane targeting of GRK2 and further the interaction of GRK2 with activated receptors. This leads to an enhanced GRK2 activity towards B2-adrenergic receptors [33]. C-Src activates GRK2: c-Src, a tyrosine kinase has been shown to directly phosphorylate GRK2 under in vitro conditions as well as it promotes tyrosine phosphorylation of GRK2 upon stimulation of B2-adrenergic receptors [34, 35]. This process is however dependent on the ability of B-arrestins to recruit c-Src since B- arrestin mutants defective in c-Src binding are defective in this process. [36]. Tyrosine phosphorylation of GRK2 directly enhances its catalytic activity as is seen by its 15 increased activity towards both soluble and membrane bound substrates. Hence, recruitment of c-Src by B-arrestins to the activated GPCR results in GRK2 phosphorylation at tyrosine residues and increases its catalytic activity. Extracellular signal regulated kinase 1 (ERKl) inhibits GRK2: ERKI, a MAPK, has also been shown to phosphorylate GRK2 at serine 670 under in vitro and in situ conditions [37, 38]. This phosphorylation site is present within the GBy binding domain of GRK2. Hence, ERKl phosphorylation of this site impairs the interaction of GRK2 with GBy, inhibiting GRK2 translocation to the activated membrane receptors thus inhibiting its catalytic activity. This process triggers a negative feedback loop that prevents accumulation of an active pool of GRK2. Moreover, both kinases have been found to co-immunoprecipitate in an agonist dependent manner [3 8]. 2.1.4.4 Regulation of GRK5 by GBy subunits and phospholipids GRK2 and GRK3 contain a PH domain (residues 561-655) in the carboxy- terminal region that partially overlaps with the GBy-binding region. Free GBy subunits bind to GRK2 with high affinity, and is required in reconstituted systems for association of GRK2 to lipid vesicles and GPCR phosphorylation [21]. This is important since both GRK2 and GRK3 are cytoplasmic proteins that transiently translocate to the membrane upon GPCR stimulation. Hence, it is suggested that the interaction of GBy with GRK2 and GRK3 targets these kinases to membrane sites where receptors are activated and GBy dimers are released. GBy binding increases the activity of GRK2 by promoting GPCR-mediated allosteric activation of GRK2. 16 PH domains of GRK2 and GRK3 can also interact directly with PIP2 and other acidic phospholipids, affecting their kinase activity [22, 39]. GRK2 phosphorylation of membrane receptors is enhanced by atleast two to three fold by binding of phosphatidyl serine (PS) and PIPZ. Among these, PS directly affects the catalytic activity of GRK2 since PS also enhances the phosphorylation of soluble substrates. PIP2 appears to bind to the amino-terminus of the PH domain suggesting that GBy and lipids have a synergistic role in GRK2 translocation and activation. GRK5 is also regulated by lipids despite lacking the PH domain. GRK5 possesses two different regions rich in positively charged amino acids (amino terminal residues 22-29 and carboxyl terminal residues 547-560) that can bind to lipids [40]. The amino terminal binding site exhibits a high specificity for PIP2 in contrast to the carboxyl terminal binding site which does not exhibit a stringent specificity for binding to lipids. PIP2 binding onto the amino terminal binding site increases GRK5 phosphorylation of receptor substrates but does not affect phosphorylation of soluble peptides or GRK5 autophosphorylation. On the other hand, lipid binding onto the carboxyl-terminal binding site stimulates autophosphorylation of GRK5 and its activity towards different substrates. These lipid binding domains are highly conserved in the GRK4 subfamily and binding of PIPZ enhances receptor phosphorylation by GRK4 and GRK6 as well. 2.1.4.5 Regulation of GRK3 by interaction with agonist activated GPCRs The activity of GRKs is also regulated by interaction with agonist stimulated GPCRs. Activity of GRKI towards a peptide substrate increases in the presence of activated rhodopsin receptor. This is because the association of GRKl with the third 17 intracellular loop of rhodopsin causes kinase activation [41]. Similarly, the catalytic activity of GRK2 is regulated in the presence of either agonist activated B2-AR and rhodopsin, or synthetic peptides derived from intracellular loops. These interactions appear to occur through the N—terminal region of GRKs [42]. It is thought that interaction of the GRKs with the receptor causes a conformational change in the GRK5 thus releasing an autoinhibitory constraint and causing kinase activation. GRK2 and GRK3 has also been shown to interact to activated Gaq subunits with high affinity, leading to a reduced Gaq mediated phospholipase C-B activation both in vitro and in intact cells [43, 44]. Hence the interaction of GRKs with the GPCRs and Gaq subunits may serve two functions, 1) translocation of GRKs to the membrane and 2) phosphorylation independent termination of signal transduction mediated by GPCRs by interfering with the binding of stimulated GPCR and Gaq with their effectors [45]. 2.1.5: Other novel interactions of GRK5 and their physiological effects Over the past few years it has become evident that the role of GRK3 is not limited to GPCR phosphorylation, desensitization and internalization. Apart from these classical functions, GRKs have now been shown to interact to non GPCR receptor substrates and to a variety of signaling proteins, regulating cell signaling in a phosphorylation dependent as well as independent manner. In this regard, GRKs have been shown to phosphorylate other non GPCR membrane receptors such as Platelet derived growth factor (PDGF) receptor [46] as well as non receptor substrates such as tubulin [47, 48], synnucleins [49], phosducin [50], ribosomal protein P2 [51], ezrin- 18 radixin-moesin (ERM) family protein ezrin [52] and NFKBI P105 [1]. In addition, GRK5 have been shown to regulate several signaling pathways in a phosphorylation independent manner by interacting with a variety of signaling as well as trafficking proteins. The novel interactions of GRKs along with their functional consequencies are summarized: PDGFRB receptor: The platelet-derived growth factor receptor-B (PDGFRB) plays a crucial role in regulating the proliferation and survival of mesenchymal cells. It has been found that GRK2 phosphorylates PDGF RB receptor on Ser1104 which is responsible for its desensitization in physiological systems. GRK2 mediated phosphorylation of Ser1104 on PDGFRB promotes its dissociation from the PDZ + + domain containing NHERF protein (Na /H exchange regulatory factor) which is involved in potentiating PDGF RB dimerization and activation, thus causing receptor desensitization [46]. Tubulin: In a quest to search for other proteins that might interact with GRK2, a GST fusion protein containing the C-terminus of GRK2 was used. GRK2 was found to bind to the cytoskeletol protein tubulin through its carboxy terminal domain (residues 467-689) by this method. Furthermore, GRK2 can phosphorylate tubulin and phosphorylation of rhodopsin is inhibited by tubulin in a dose dependent manner [47]. However, the physiological relevance of this interaction / phosphorylation is not very clear yet although it is possible that the GRK2-tubulin interaction might have a role in regulating the desensitization of GPCRs by GRK2. Furthermore, it is also speculated that GRK5 might play a role in regulating microtubule dynamics and cytoskeletol reorganization. 19 Synuclein: To identify new substrates for GRK3, bovine calf brain extracts were prepared and were used in phosphorylation reactions. Synuclein, a protein highly expressed in brain was identified as a substrate by this method. Lipids and GBy subunits, which stimulate phosphorylation of GPCRs by GRK3, also activate synuclein phosphorylation by GRK3. Furthermore, all GRKs can phosphorylate synuclein. Phosphorylation of synuclein by GRKs blocks the interaction of synuclein with phospholipids. This in turn reduces the inhibitory effect on Phospholipase D2 (PLD2), an enzyme involved in regulating vesicular trafficking. Hence, it is possible that agonist bound GPCRs activate GRKs which then phosphoryate GPCRs targeting them for endocytosis as well as also phosphorylate synuclein thus activating PLD2 which is involved in vesicle formation needed for endocyotsis as well as recycling of receptors. Raf kinase inhibitor protein (RKIP): RKIP is a protein that belongs to a family of phosphatidylethanolamine-binding proteins (PEBPs) that serve as inhibitors of kinase signaling pathways. RKIP is a physiological inhibitor of Ras-Raf-MEK- ERK pathway involved in several cellular processes such as differentiation, proliferation, cell survival and cell transformation. RKIP has been shown to interact with GRK2 [53]. GPCR stimulation leads to the activation of PKC which can phosphorylate RKIP on serine 153. Phosphorylation of RKIP leads to an increase in its affinity towards GRK2 which then leads to its dissociation from its target Rafl, prolonging the activation of ERK kinase. The interaction of GRK2 with RKIP blocks the kinase activity of GRK2, impairing the desensitization process, and leading to a prolonged signaling. 20 Phosphoinositide 3-kinase (PI3K): PI3Ks are a conserved family of lipid kinases whose function is to catalyze the addition of a phosphate on the third position of inositol ring. Stimulation of GPCRs causes activation of PI3Ks increasing the levels of D-3 phosphatidyl inositols which are potent signaling molecules. A direct protein- protein interaction has been shown for PI3Ky and GRK2 through 197 amino acid residues PIK domain in PI3K [54]. GRK2 and PI3Ky form a cytosolic complex and translocate to the membrane in an agonist dependent manner. This interaction plays an important role in receptor sequestration as it has been shown that inhibition of kinase activity of PI3K results in attenuation of B-adrenergic receptor sequestration. Furthermore, cardiac specific overexpression of the PIK domain causes disruption of the GRK2-PI3K interaction under in vivo conditions, preserving B-adrenergic receptor signaling even after prolonged catecholamine administration. This helps restore myocardial contractibility to normal in heart failure conditions [55]. Akt: Akt is a serine threonine kinase which directly interacts with GRK2. The region of interaction has been determined to be GRK2 C-terminus (aa 492-689). This interaction leads to an inhibition of Akt phosphorylation. A study using rats with portal hypertension showed an enhanced expression of GRK2 in their sinusoidal endothelial cells. Enhanced GRK2 was found to lead to an inhibition of Akt phosphorylation. Since Akt phosphorylation is required for the production of NO by eNOS, inhibition of NO production was found to lead to portal hypotension. [56]. GPCR-kinase interacting proteins (GIT proteins): The GIT family of proteins were found to be binding partners of GRKs [57]. Their structure consists of a zinc-finger motif and several ankyrin repeats in the N-terminal portion with a GAP 21 (GTPase activating protein) domain in the first 45 amino acids. The GIT proteins are active as GAPS on ARF 6 protein present on the plasma membrane. Yeast two hybrid screen has identified GITl and GIT2 as binding partners for GRK2, 3, 5 and GRK6 [57]. However, the functional significance of this interaction is not very clear as yet. MEK: GRK2 and MEKl has also been shown to be present in the same multimolecular complex. Furthermore, it was found that elevated levels of GRK2 inhibit chemokine mediated induction of ERK whereas decreased levels of GRK2 promote chemokine mediated ERK activity. MEK activity was not found to be affected though. Furthermore, neither the kinase activity nor the interaction of GRK2 to G protein subunits is necessary for this effect. [58]. Heat shock protein 90 (Hsp 90): Hsp 90 is a highly conserved chaperone protein that interacts with a wide variety of signaling proteins. GRK2 has been found to interact with Hsp 90 and it has been shown that the disruption of this interaction causes an increased degradation of GRK2 via the proteasome pathway. This suggests that the interaction of GRKs with the heat shock proteins plays an important role in the folding and maturation of GRKs [59]. 2.2 Physiological and Pathological roles of GRKs Development of genetically modified mouse models with either a targeted deletion of a particular GRK or overexpressing a GRK transgene has given us a great insight into the roles of individual GRKs in various signaling pathways as well as in an intact animal. 22 2.2.1: Physiological roles of GRKs as determined by knockout / transgenic mice GRK] is required for rhodopin desensitization: GRKl or rhodopsin kinase was the first GRK to be discovered [60] and was found to phosphorylate light activated rhodopsin at C-terminal residues. It is expressed in retina and in mammalian pineal gland. A better idea of the role of GRKl under in vivo conditions came with the -/- . generation of mice deficient in GRKl (GRKI ) [61]. Deficrency of GRK] led to an elimination of the light dependent phosphorylation of rhodopsin causing the single photon response to become larger and longer than normal. Also, a day of constant light caused the rods in these mice to undergo apoptotic degeneration. Furthermore, the cone response recovered around 30-50 times slower than normal when the mice were exposed to a bright conditioning flash. GRK2 plays an essential role in embryonic development: Similar to GRKl, studies with GRK2 knockout mice confirmed an important role of GRK2 in . . , -/- embryonic cardiac development and function. GRK2 (GRK2 ) homozygous knockout mice embryos die during gestation. Examination of these embryos revealed that there was a pronounced hypoplasia of the ventricular myocardium as well of the interventricular septum due to which the atrial and ventricular cavities appeared to be unusually large [62]. Furthermore, it was found that there was a 70% decrease in left ventricular ejection fraction suggesting an impaired heart function. These mice, however, developed normally with no apparent adult cardiac phenotype at baseline except a modestly enhanced contractile function when GRK2 was specifically deleted in cardiac myocytes [63]. This study suggests that GRK2 does not play a specific role 23 in cardiac development, rather it might have a much more broader role in embryonic development. A specific role for GRK2 in embryonic development has not been identified yet though. Contractile response to B-adrenergic receptor and angiotensin II receptor stimulation is attenuated by myocardical overexpression of GRK2 [64]. Vascular smooth muscle (VSM) specific overexpression of GRK2 similarly attenuates B-adrenergic receptor signaling resulting in increased resting blood pressure [65]. GRK3 knockout mice exhibit supersensitivity to olfactory stimuli: GRK3 knockout mice have a normal embryonic as well as postnatal development. GRK3 . -/- . . deletion (GRK3 ) has shown that GRK3 plays a role in the regulation of olfactory . . . -/- . . . receptors. Cilia preparations from GRK3 mice do not show a fast agonist induced desensitization which is normally seen after stimulation with odorants [66]. GRK4 regulates dopamine-l receptor: Transgenic mice overexpressing GRK4 were generated whereby it was found that the transgenic mice overexpressing wild type GRK4y showed a normal phenotype in contrast to the mice carrying a naturally occurring polymorphism A142V in GRK4y which showed increased kinase activity and hypertension. The diuretic and natriuretic effects of D1 agonist were also found to be impaired in these mice suggesting that GRK4 regulates dopamine-1 receptor in kidney [67]. GRK5 is required for M2 muscarinic receptor desensitization: GRK5 . -/- . . . . . deletion (GRK5 ) caused an increase in cholrnergic responses such as hypothermia, hypoactivity, tremor, salivation and antinociception [68]. Furthermore, central M2 muscarinic receptors have been found to be resistant to desensitization in the absence 24 of GRK5. It was also found that B2-adrenergic receptor induced airway smooth . . -/- . . . . . muscle relaxation was reduced in GRK5 mice [69]. Transgenic mice wrth cardiac specific overexpression of GRK5 show enhanced B-adrenergic receptor desensitization in comparison to non-transgenic control mice. However, there was no change in the contractile response to angiotensin II receptor stimulation [70]. Vascular smooth muscle (VSM) specific overexpression of GRK5 showed an increase in blood pressure which was also found to be gender dependent i.e. male mice showed a much larger increase in blood pressure as compared to that in female mice [71]. However, unlike the VSM specific overexpression of GRK2, overexpression of GRK5 in VSM did not cause vascular or cardiac hypertrophy. GRK6 plays an essential role in desensitization of chemokine receptors: T cells from GRK6 knockout mice (GRK6’l') show an impaired chemotactic response to CXCL12 (ligand for CXC chemokine receptor 4 CXCR4) [72] whereas neutrophils from the same mice showed an enhanced chemotactic response to CXCL12 [73] as well as to leukotriene B4. Furthermore, acute migration of neutrophils from the bone marrow in response to G—CSF has been found to be impaired suggesting that this might be due to CXCR4-mediated retention of neutrophils in the bone marrow [73] (Table 2.2). 2.2.2: Pathological roles of GRK5 Change in GRK5 expression and activity have been found in several pathologies suggesting an important role of GRKs in particular disease conditions. Reduced response to B-receptor agonist and a loss of cardiac contractility in human 25 chronic heart failure has been shown to be linked to an increased expression of GRK2 and reduced expression of B1 receptor [74, 75]. Furthermore, the activity as well as the expression of GRK2 in lymphocytes is significantly enhanced in hypertensive subjects as compared to normotensive controls [76]. This increase negatively correlates with B- agonist stimulated lymphocyte adenylyl cyclase activity [77]. Reduced adenylyl cyclase activity correlates with a reduced vasodilator response to B-adrenergic agonist stimulation of vascular smooth muscle receptors in hypertensive state [78]. A significant decrease in expression as well as activity of GRK2 and GRK6 has been shown in peripheral blood mononuclear cells (PBMCs) of patients with rheumatoid arthritis as compared to those from healthy individuals [79]. This reduced expression and activity of GRKs correlates with an increased sensitivity to B2- adrenergic stimulation as B-agonist induced CAMP production has also been found to be increased in leucocytes from these patients. Furthermore, it was found that beta blockers and receptor antagonists for a chemokine, MCPl (Monocyte Chemoattractant Protein 1) can reduce the severity of disease in chronic arthritis [80, 81]. This is important since chemokine receptors which fall under classical GPCRs, as well as other GPCRs such as those for prostaglandins and substance P play an important role in inflammation in Rheumatoid arthritis. This suggests that cell type specific decrease in GRKs activity might be responsible for sustained activation of G—protein coupled pro-inflammatory receptors and hence defects in cellular metabolism. Rat models of 26 Table 2.2: Phenotypic characteristics of GRK Knockout/Transgenic mice Modified from: Premont, RT and Gainetdinov, RR. Physiological roles of G-protein coupled receptor kinases and arrestins. Annual Review of Physiology 2007 69:511-534 Metaye, T., Gibelin, H., Perdrisot, R., Kraimps, GL. Pathophysiological roles of G-protein coupled receptor kinases. Cellular Signaling 2005 17:917-928 27 Knockout Phenotype GRKl Prolonged response and light-induced apoptosis in rods Slow resensitization in cones GRK2 double allele deletion GRK2 single allele deletion GRK2 cardiac specific deletion VSM specific overexpression of GRK2 VSM specific deletion of GRK2 GRK2 ablation in myeloid cells Embryonic lethal, thin myocardium syndrome in embryos Enhanced myocardial contractile response to a B- agonist Enhanced chemotaxis of T-lymphocytes Altered progression of experimental autoimmune encephalomyelitis Enhanced inotropic sensitivity to isoproterenol VSM BAR signaling attenuated Diminished BAR mediated dilation High blood pressure Enhanced BAR mediated dilation Does not rescue high blood pressure Increased an) AR stimulation Earlier onset of H1 (Hypoxia-Ischemia) brain injury GRK5 myocardial overexpression GRK3 Enhanced airway response to a cholinergic agonist Loss of kappa opioid tolerance Lack of olfactory receptor desensitization GRK4 Normal fertility and sperm function. No obvious phenotype GRK5 Excessive opposition of airway smooth muscle relaxation after B—agonist administration Enhanced hypothermia, hypoactivity, tremor, and salivation by oxotremorine Attenuation of contractility and heart rate in response to a B—agonist _G_RK6 Enhanced locomoter- stimulating effect of cocaine and amphetamine Impaired chemotaxis of T-lymphocytes Enhanced chemotaxis of bone-marrow-derived neutrophils and impaired acute neutrophil mobilization in response to G-CSF Altered central dopamine receptor regulation 28 arthritis also showed that changes in GRK2 and GRK6 levels occur after induction of arthritis. GRK2 has also been found to be reduced in platelets from patients with depression whereby treatment with antidepressants has been found to cause an upregulation in GRK2 levels [82]. Lung homogenates from Cystic fibrosis patients show a decreased B-agonist stimulated adenylyl cyclase activity and a simultaneous increase in GRK activity as well as mRNA and protein for both GRK2 and GRK5 [83] suggesting that an increase in GRKs might be responsible for alteration in B2- adrenergic receptor density. 2.3: GRKs and inflammation GRKs play an important role in inflammation and disease. Firstly, GRK2, 3, 5 and 6 have been found to be expressed at particularly high levels in immune cells [84] and their levels in immune cells are dynamically regulated in response to inflammation suggesting an important role for these kinases in immune activity. Secondly, targeted deletion of GRKs in vivo affects the progression of various acute as well as chronic inflammatory diseases again suggesting an important role for GRKs in disease. Furthermore, chemokine receptor signaling has also been shown to be tightly regulated by GRKs under in vitro conditions suggesting that GRKs play an important role in inflammation. 2.3.1: GRKs expression and inflammation: Mak et al., showed an increased expression of GRK2 and GRK5 in the lungs of rats treated with IL-IB, a pro- 29 inflammatory cytokine, and this effect was found to be abolished when the rats were treated with an anti-inflammatory steroid, dexamethasone [85]. Pro-inflammatory cytokines as well as oxygen radicals have also been found to reduce the levels of GRK2 under in vitro conditions [79, 86]. LPS has been shown to downregulate chemokine induced expression of GRK2 and GRK5 in neutrophils (PMNs) [87]. Moreover, pro-inflammatory cytokines such as IL-1, TNF-a and IFN-y regulate GRK2 expression at the transcriptional level by regulating the GRK2 promoter [88]. In one study, our lab found that stimulation of mouse peritoneal macrophages with ligands for TLR2, TLR3, TLR4 and TLR7 caused an upregulation in the levels of GRK2. Conversely, stimulation of mouse peritoneal macrophages with ligands for TLR2, TLR4 and TLR7 caused a significant decrease in the GRK5 and GRK6 protein levels [89]. In another study, it was found that Lipoteichoic acid (LTA), a known TLR2 agonist, increased the expression of GRK2 in neutrophils. This corresponded with a downregulation in the expression of chemokine receptor CXCR2 in neutrophils in, sepsis and an impaired migration of neutrophils into an infectious focus in vivo and reduced chemotaxis in vitro [90]. These studies suggested that GRKs have an important role in inflammation since TLRs are important mediators of inflammatory response. 2.3.2: GRKs deletion and inflammation: Heterozygous GRK2 knockout mice . +/- (GRK2 ) have been found to have an advanced onset of multiple sclerosis with increased numbers of inflammatory cells in the spinal cord as compared to control mice [91]. However, overexpression of GRK2 in vascular smooth muscle cells has 30 been found to induce hypertension and cardiac hypertrophy [65]. Another finding shows that arachidonic acid induced acute ear inflammation but not PMA-induced . -/- —/+ . . inflammation increased substantially in GRK6 and GRK6 mice suggesting a specificity towards an agent causing inflammation rather than a general effect of GRK6 deficiency on the chemotactic activity of neutrophils [92]. 2.3.3: GRKs regulation of chemokine signaling and inflammation: GRKs can also regulate chemokine responses in inflammation. Increased signaling by chemokine receptors plays an important role in disease pathology, regulating the migration of . +/- . neutrophils, monocytes and T-cells. In this regard, T-cells from GRK2 mice have been found to have enhanced CCRS agonist induced calcium mobilization, PKB and ERK1/2 signaling and migration towards CCRS agonist under in vitro conditions [93] + Spleenocytes from GRK2 mice were also found to have an enhanced CCR2 mediated signaling to ERKl/Z [5 8]. Hence, it is clear that the alterations in expression and activity of GRKs as well as the role of GRKs in regulating chemokine mediated responses play an important role in inflammatory diseases and should be taken into account when developing therapeutic strategies for such conditions (Table 2.3). 2.4: LPS and TNFa induced inflammation 2.4.1 LPS induced inflammation 31 Table 2.3: Involvement of GRKs in diseases Slightly modified from Metaye, T., Gibelin, H., Perdrisot, R., and Kraimps, IL (2005) Cellular signaling 17(8), 927-928 32 GRK Disease GRK modification GRKI Oguchi disease Genetic mutation associated with a loss of GRK activity GRK2 Chronic heart Increased expression of mRNA and protein failure GRK4y Hypertension Genetic variants with a marked stimulation of GRK4]! activity GRK2 Hypertension Increased protein expression GRK2/GRK6 Rheumatoid Decreased protein expression arthritis GRK2/GRK6 Opiate addiction Decreased protein expression GRK2 Differentiated Increased enzymatic activity thyroid carcinoma GRK2/GRK4y/5 Ovarian cancer Increased protein expression GRK3 Hyperfunctioning Increased protein expression thyroid nodule GRK2 Depression Increase in membrane fixation GRK3 Bipolar disorder Single nucleotide polymorphism in the promoter region of GRK3 GRK2/GRK5 Cystic fibrosis Increased expression of mRNA and protein GRK2/GRK3/ Left ventricular Increased expression of mRNA and protein GRK5 overload diseases GRK2/GRK6 Cardiopulmonary Decreased protein expression bypass in cardiac diseases 33 The mammalian innate immune system defends against bacterial or viral infection and essentially forms the first line of host defense. Toll like receptors (TLR) are crucial components of the innate immune system, and recognize a wide variety of microbial products known as pathogen associated molecular patterns (PAMPs) present in bacteria, viruses and other pathogens. Of the 10 human TLRs, the best characterized is TLR4, which recognizes gram negative bacterial outer membrane structural component known as lipopolysaccaride (LPS) activating immune cells such as monocytes and macrophages leading to the induction of endotoxic shock in mammals. 2.4.1.1: Toll like receptor 4 (TLR4) signaling TLR4 works in concert with a coreceptor protein CD14 plus a secreted protein MD2 to transmit cell signals whereby CD14 is required for presentation of LPS to TLR4-MD2 [94]. Formation of LPS—TLR4-MD2 complex is required for the activation of downstream signals. LPS recognition causes TLR4 to undergo oligomerization and recruitment of downstream adapter proteins through interaction with the TIR domain (Toll-interleukin-l receptor). The TIR domain is highly critical for signal transduction since it has been found that a single point mutation in the TIR domain can abolish the response to LPS [95]. TLR4 utilizes five TIR domain containing adapter proteins. These are MyD88 (Myeloid differentiation primary response gene 88), TIRAP (TIR domain-containing adapter protein, also known as Mal or MyD88-adapter-like), TRIF (TIR domain- containing adapter inducing [FN-B), TRAM (TRIP-related adapter molecule), and SARM (Sterile alpha and HEAT- arrnadillo motifs-containing protein) [96]. 34 TLR4 signaling has been divided into MyD88 dependent and MyD88- independent (TRIP-dependent) pathways. Under MyD88 dependent signaling, upon LPS stimulation, MyD88 activates a death domain-containing kinase IRAK-4 (IL-1 receptor-associated kinase-4). IRAK-4 leads to the activation of TRAF6 (TNF receptor associated factor 6). TRAF6 activates TAKl (Transforming growth factor-B- activated kinase 1) which then activates IKK (IKB kinase) and MAPK pathways [97, 98]. IKB kinase finally leads to the activation of NFKB which controls the expression of pro—inflammatory cytokines and chemokines as well as various other genes involved in innate and adaptive immunity. Activation of MAPK causes induction of AP-l, another transcription factor which is also involved in the expression of pro- inflammatory cytokines and chemokines [99]. It has been found that NFKB and MAPK are activated through MyD88 independent pathway as well. MyD88 independent signaling is carried on by TRIF which is another TIR- domain containing protein. MyD88-independent pathway also leads to the activation of NFKB, however, this activation occurs later than the MyD88-dependent activation. It was shown by Covert et al. that NFKB activation by MyD88-independent pathway requires protein synthesis which leads to a delay in activation. It was later discovered that TRIP-dependent pathway activates TNFa expression and secretion in an NFKB- independent manner through a TRIP-dependent pathway specific transcription factor IRF3. The secreted TNFa then binds onto its receptors and activates NFKB. This SUggested that the activation of NFKB by TRIP-dependent pathway occurs by a secondary response through TNFa, thus resulting in an autocrine pathway for delayed NFKB activation [100] (Fig. 2.2)- 35 LPS challenge leads to a quick production of TNFa peaking at 1.5 hours. Excessive production of pro-inflammatory cytokines such as TNFa not only enhances immune responses required for fighting against invading pathogens but can also produce deleterious effects that perturb regular hemodynarnic and metabolic balances in the system. TNFa is produced in very high levels during the early phase of the response to LPS induced endotoxemia. TNFa in turn can affect the expression levels of TLR4 suggesting some feedback regulation [101]. 2.4.2 TNFa-a potent pro-inflammatory cytokine Tumor necrosis factor-alpha (TNFa) is a pro-inflammatory cytokine primarily secreted by activated macrophages and monocytes. It is expressed as a 26kDa type II membrane-bound protein that self-associates into a bioactive homotrimer [102, 103] Normally TNFa is kept in balance by other anti-inflammatory factors. But in case of inflammation, this balance is shifted and TNFa levels increase which in turn cause upregulation of adhesion molecules on the endothelium (such as ICAMl and VCAMI) and cause stimulation of fibroblasts leading to their proliferation, and recruitment of leukocytes to the site of inflammation. TNFa also stimulates the production and release of other cytokines and chemokines from macrophages. TNFa mediates its diverse effects through two cell surface receptors: p55TNFR1 and p75TNFR2. TNFRl is constitutively expressed on almost all nucleated cells whereas TNFR2 is expressed mainly on immune and endothelial cells [104]. The TNFRl contains a death domain (DD) which is lacking in TNFR2 [105]. 36 Figure 2.2: LPS activates NFKB and MAPK pathways Stimulation of TLR4 receptor by LPS causes TLR4 to dimerize and recruit MyD88. Myd88 binds IRAK4 which in turn phosphorylates IRAKl. IRAKI binds TRAF6 which in turn binds a preformed complex bound to the membrane consisting of TABl/TAKl/TABZ. Active TAKl then phosphorylates and activates the IKK complex which in turn phosphorylates and activates IKBa which undergoes proteasomal degradation releasing NFKB subunits (p50 and p65) which then translocate to the nucleus to initiate gene transcription. LPS also activates MAP kinases namely c-Jun N-terminal kinase (INK), p38 and ERK1/2 MAP kinase. In macrophages, however, stimulation with LPS leads to H(KB mediated phosphorylation and degradation of NFicBl p105 releasing associated p50 subunit which translocates to the nucleus to modulate gene transcription. p105 degradation also liberates the associated MAP3 kinase (TPL-2) which then activates ERK kinase. “Images in this dissertation are presented in color”. 37 .i ,1 j,_P\b p Proteasomal degradation NFKB target . - ene transcription Transcription 38 Figure 2.3: TNFa stimulates NFKB and MAPK pathways Binding of TNFa onto TNF receptor causes receptor trimerization, which facilitates recruitment of adapter protein TRADD. TRADD leads to the recruitment of RIPl to the signaling complex. RIP] acts as an adaptor that facilitates recruitment of IKK complex to TNFR. H(KB then phosphorylates and activates IKBa which undergoes proteasomal degradation releasing NFKB subunits (p50 and p65) which then translocate to the nucleus to initiate gene transcription. TNFa also activates MAP kinases namely c—Jun N-terminal kinase (JNK), p38 and ERKl/2 MAP kinase. In macrophages, however, stimulation with TNFa leads to IKKB mediated phosphorylation and degradation of NFKBl p105 releasing associated p50 subunit which translocates to the nucleus to modulate gene transcription. p105 degradation also liberates the associated MAP3 kinase (TPL-2) which then activates ERK kinase. “Images in this dissertation are presented in color”. 39 Proteasomal degradation » NFKB target gene transcription Transcription 40 Stimulation of TNFRI leads to the recruitment of DD-containing adapter molecule, Tumor necrosis factor receptor 1 associated death domain protein (TRADD) [106] followed by binding of DD-containing Ser/Thr kinase, Receptor interacting protein-1 (RIPl) [107]. This signaling complex is then bound by other adapter proteins such as TNF receptor associated factor 2/5 (TRAF2/5) and Inhibitor of apoptosis 1 (c- IAPl) which subsequently leads to the activation of NFKB and MAPK pathways (Fig. 2.3) [108]. 2.5: NFKB transcription factor in inflammation NFKB regulates transcription of genes involved in inflammation, the innate and adaptive immune responses, cell proliferation, cell adhesion, genes involved in controlling programmed cell death (apoptosis), and genes involved in cellular stress response and tissUe remodeling [109-113]. It was discovered as a transcription factor that binds to the intronic enhancer of the kappa light chain gene (the KB site) in B cells around 20 years ago. The NFKB family of transcription factors consists of five members viz. NFKB-l (p105 precursor and p50), NFKBZ (p100 precursor and p52), Rel A (p65), c-Rel and Rel B. All of these possess a Rel homology domain (RHD) at the N-terminus which is required for homo and hetero dimerization as well as for sequence specific DNA binding. Rel A, Rel B and cRel also contain a transcription activation domain (TAD) at their C-terminus which is absent in the p50 and p52 subunits. Hence, p50 and p52 interact with other factors to positively regulate transcription. NF KB is sequestered in the cytoplasm in the form of an inactive complex with IKB family of inhibitory protein under unstimulated conditions [114]. The IKB 41 family consists of three classical members viz. IKBa, IKBB, and IicBe all of which are characterized by the presence of multiple ankyrin repeats that are important for binding to NFKB dimers and interfere with the nuclear localization signals present in the NFKB. NFicBl (p105) and NFKBZ (p100) are synthesized as large precursors that contain RHD at their N-terminal and multiple ankyrin repeats in their C-terminal halves due to which they function as IKB like proteins. Proteolysis of the C-terminus of these precursors yields mature P50 and P52 subunits [115]. Classical or canonical pathway of NFKB activation typically involves IKKB (subunit of IKB kinase complex)-dependent phosphorylation and subsequent 26S proteasomal degradation of IKBa, IKBB, and 168. The kinetics of phosphorylation and degradation of IKBB and IKBe are however much slower than that of IKBO. (Fig. 2.4). The proteasomal degradation leads to the release and translocation of NFKB (most commonly the P50-Rel A dimer) to the nucleus [116] where it causes an increased transcription of genes encoding for cytokines, chemokines, adhesion molecules (ICAMl or Intercellular adhesion molecule 1, VCAMl or vascular cell adhesion molecule 1, ELAM or endothelial leucocyte adhesion molecule 1), matrix metalloproteinases (MMPs), cyclo-oxygenase 2 (COX2), inducible nitric oxide synthase (iNOS) as well as inhibitors of apoptosis all of which are important part of an innate immune response [117]. As further elaborated in chapters 3 and 4, our studies both in cultured cell line as well as primary cells implicate GRK5 as an important mediator of NFKB pathway by potentially phosphorylating the same residues as that of IKKB. These results suggest that GRK5 might be a better and alternative target to 42 Figure 2.4: Mammalian NFKB and IKB family members The Nuclear factor KB (NFKB) family has five members. These are Rel A (P65), c-Rel, Rel B, p105/p50 (NFKBI) and p100/p52 (NF KB2). At the amino terminal, there is a structurally conserved Rel homology domain (RHD). RHD contains the dimerization, nuclear localization (N) and DNA-binding domains. Rel A, Rel B and c-Rel also have a carboxy terminal non homologous transactivation domain (TAD). Rel B also contains a leucine zipper motif (LZ). Inhibitor of NFKB (IKB) family has IKBa, IKBB, IKBe and Bcl3 as members all of which have multiple ankyrin repeats (ANK). p105 and p100 contain RHDs at the amino terminus and ANK repeats at the carboxyl-terminus due to which they act like IKB proteins. P105 and p100 undergo proteolytic processing to generate p50 and p52 as NFKB proteins. The number of amino acids in each protein are shown on the right. “Images in this dissertation are presented in color”. 43 D11 257 mini mum HE; A Z «38:. , ALE! Emil .r _ _ $323 xz<2m~ .. afiiw E M view”. 9.me ad; a. H E 31.0 ear 9: .3 m :33. 44 that of IKKB, since IKBO. phosphorylation and NFkB activation is only partially affected. However, there is a more recently discovered alternative pathway of NFKB activation which involves IKKa (subunit of IKB kinase complex)—dependent phosphorylation and degradation of P100 and subsequent activation of p52:RelB dimers [118]. Furthermore, IKKB dependent degradation of IKBa has been found to occur within minutes whereas the kinetics of IKKo. dependent degradation of p100 is slower and requires several hours. IKBa is the best studied IKB family member and is known to regulate the classical RelAszO heterodimers. In response to inflammatory stimuli such as LPS or TNFa, it undergoes rapid degradation and is finally resynthesized in an NFKB dependent fashion which constitutes a negative feedback loop whereby newly synthesized IKB enters the nucleus and associates with deacetylated RelAszO heterodimers shuttling them back to the cytoplasm [119, 120]. LPS and TNFa act as important stimulators of NFKB signaling pathway. NFKB transcription factor is activated in tissues such as liver, lungs and spleen within 4 hours of intra-peritoneal challenge of LPS [121]. It activates more than 150 genes, most of which are pro—inflammatory in function. Deficient NFKB activation in intestinal epithelium has been linked to an increased inflammation under in vivo conditions suggesting that a defect in NFKB signaling can lead to immunosuppression which triggers and also maintains inflammation [122]. NFKB has been found to be highly activated in several inflammatory disease conditions such as sepsis, rheumatic diseases, inflammatory bowel disease, multiple 45 sclerosis etc. as well as in other diseases such as cancer and diabetes as detected by electrophoretic mobility shift assays as well as tissue staining using NFKB specific antibodies of biopsies from these patients. These changes are in turn accompanied by an increased recruitment of inflammatory cells to the tissues. Hence, although a prompt activation of NFKB is required for a good immune response, it should also be terminated properly to prevent from reaching a stage of tissue damage, organ failure and finally death. Inhibition of NFKB activation has been shown by several studies to be effective in controlling inflammatory disease conditions [123]. Several drugs used to treat inflammatory disease conditions have effects on NFKB activity. Anti TNFa drugs have been found to be highly effective in controlling inflammation and are widely used against rheumatoid arthritis. Moreover, anti-inflammatory drugs such as corticosteroids, aspirin and other non-steroidal anti-inflammatory drugs (NSAIDS) although do not directly target NFKB, they do control the expression of genes regulated by NFKB [124]. In sepsis, inhibition of NFKB activation prevents LPS-induced iNOS and COX2 mRNA and protein expression and activity. NFKB inhibition also corrects cardiovascular functional abnormalities as well as help restore systemic hypotension in sepsis [125]. Inhibition of IKK activity also reduces cytokine production and decreases infiltration of neutrophils in lungs, liver and colon thus improving the rate of survival in polymicrobial model of sepsis [126]. Inhibition of NFKB activation further reduces LPS induced increases in microvascular permeability. In liver, inhibition of NFKB activation has been found to suppress LPS induced cytokine and adhesion 46 molecule expression, reduce influx of neutrophils as well as prevent LPS induced increase in microvascular endothelial permeability [127]. Although NFKB is an attractive target for therapeutic inhibition under certain disease conditions, considering its role in normal cellular physiology as well as in mounting effective immune response, its inhibition might have serious effects. Thus, although it has been suggested by several studies that inhibition of NFKB activation in viva leads to reduced inflammatory responses in sepsis, it also favors apoptosis in immune cells which may lead to immunosuppression and fatal outcome in severe sepsis. As for instance, it was found that inhibiting NFKB exacerbated acute inflammation but helped attenuate chronic inflammation in the intestinal tract. NFKB protects against epithelial cell apoptosis which was reduced leading to an aggravation of an acute inflammatory response. In chronic inflammation however the risk of epithelial cell apoptosis is absent, thus NFKB inhibition is beneficial under such conditions [128]. In another study, it was found that mice with a targeted deletion of IKKB in myeloid cells are more susceptible to endotoxic shock due to an increased production of plasma IL-lB suggesting the possible complications that could arise from IKKB inhibition [129]. Blockage of NFKB could also compromise normal host defenses. It was shown that mice were unable to clear opportunistic infections such as those involving Listeria monocytogenes after NFKB inhibition [130]. Another study showed that blocking NFKB pathway in LPS model of sepsis inhibited inflammatory as well as injury promoting responses thereby improving survival. On the other hand, blocking NFKB pathway in a bacterial model of sepsis inhibited host defense 47 responses in addition to inhibiting inflammatory and injury promoting responses. Thus the beneficial effects of inhibiting inflammatory responses would be compromised by the impairment of bacterial clearance capacity. Hence, the best approach would be to identify specific targets in the NFKB signaling pathway in different disease conditions for therapeutic targeting rather than considering global inhibition of NFKB. 2.6: MAPKs in inflammation Cells continuously respond to signals in the extracellular environment by sensing through cell surface receptors and further transmitting the signals to the cytosol and nucleus by activating signal transduction pathways. MAPKs constitute a family of highly conserved serine-threonine kinases that play a role in several cellular processes such as cell proliferation, cell survival/apoptosis, differentiation as well as cell stress and inflammatory conditions. 2.6.1: ERKl/Z kinase ERK1/2 is one of the three MAP kinases that is induced under inflammatory conditions by LPS as well as TNFa. Activation of ERK cascade may be responsible for monocyte and macrophage reprogramming and thus dysregulation of cytokine cascade. It has been suggested that MEK-ERK1/2 pathway is activated in a Raf-1 dependent manner by LPS as demonstrated by the inhibition of LPS induced TNFa production upon repressing Ras or Raf-l [131]. Furthermore, inhibition of MEK in monocytes causes reduction in the LPS induced production of certain pro- inflammatory cytokines such as We, IL-1, IL-8 and PGE2 which suggests that ERK plays a crucial role in inflammatory gene expression [132]. ERK inhibitors have been 48 found to be useful in reducing inflammation in an ear edema model in mice as well as experimental osteoarthritis model in rabbits [133]. In macrophages however, activation of ERK1/2 MAP kinase requires a serine- threonine kinase TPL2 (Tumor progression locus 2) also known as COT. TPL2 in turn is present in a complex with NFKBI p105. Stimulation with LPS causes phosphorylation and proteasomal mediated degradation of NF KBl p105. This releases p50 as well as TPL2. In unstimulated macrophages, TPL2 MEK kinase activity is blocked due to its association with p105. Released TPL2 fimctions as a MAP3 kinase which phosphorylates MAP2 kinase MEK1/2 which in turn phosphorylates and activates MAP kinase ERK1/2 [134]. It has been found that LPS upregulation of TNFa and COX2 is reduced in TPL2 deficient macrophages due to a defective ERKl/Z activation suggesting an important role of this pathway in mediating an immune response [135, 136]. Thus, in macrophages LPS stimulation of IKK complex plays a role in activating both NFKB and ERK pathways via NFKB] p105 regulation (Figure 2.2). Interestingly, as described in the studiesin chapter 5, GRK2 appears to be a negative regulator of this pathway in primary peritoneal macrophages. Previous studies in Raw264.7 macrophage cell line however have shown that NFKB] p105- ERK pathway is regulated by GRK5. The implications of our current findings in primary cells are further discussed in chapter 5. 2.6.2: P38 MAPK The p38 mitogen activated protein kinase is activated by diverse stimuli such as UV light, heat shock, certain mitogens, LPS as well as pro—inflammatory cytokines 49 such as IL-1 and TNFa. It has been shown that TNFa stimulation of neutrophils causes an enhanced p38-MAPK dependent phosphorylation and activation of PLA2 which is a primary regulator of arachidonate signaling [137]. Hence, p38 directly affects arachidonate signaling which can both stimulate and suppress inflammatory responses. P38 also plays a role in the production and release of cytokines such as TNFa and IL-1 [138]. P38 also plays a major role in the movement of neutrophils to inflammatory sites. P38 is involved in increasing the expression of ICAM-1 (adhesion molecule) on vascular endothelium which is required for binding of neutrophils through L-selection in the initial steps in the migratory process. Furthermore, p38 is required in the efficient generation of reactive oxygen species by NADPH oxidase activity in response to multiple agonists once the neutrophils reach their destination [139]. P38 has also been suggested to play a role in some chronic neuronal diseases such as Alzheimer’s where it is found to be activated in association with the neurofibrillary tangle-bearing neurons containing the aggregated paired helical filament protein tau [140]. P38 is also required for the angiogenesis in inflammation in a murine model of collagen induced arthritis [141]. P38 is required for the upregulation of Bl bradykinin receptors. These receptors are present at very low levels in most tissues. However, they are strongly upregulated after various types of tissue injury. It acts as a potent vasodilator resulting in increased capillary permeability which causes accumulation of extracellular water bed in conditions such as acute respiratory distress syndrome [142]. Several studies have shown the role of p38 MAPK in sepsis. In one such studies, it was shown that the p38 MAPK activity was markedly increased in splenic and peritoneal macrophages and inhibition of p38 50 MAPK markedly improves survival in a polymicrobial ceacal ligation and puncture (CLP) model of sepsis [143]. 2.6.3: JN K kinase JNK is the third important MAP kinase that is activated under inflammatory stimuli. LPS has been shown to activate JNK kinase in THPl monocytic and Raw 264.7 cells as well as other cell types. JNK kinase induces transcription factors namely AP-l, c-Jun, ATP-2, and Elk-1, all of which are important mediators of inflammatory gene transcription. JNK activation of AP-l is important for synthesis of TNFOL, as well as proliferation and differentiation of T-cells. JNK is found to be activated in joint synoviocytes suggesting an association to rheumatoid arthritis possibly due to an enhanced production of TNFOL. However, JNK deficiency has been found to reduce the progression of experimental autoimmune encephalitis (EAE), due to an increase in the expression of an anti-inflammatory cytokine IL-10 by macrophages [144]. 2.7 : Septic shock Encounter with microbes or their components such as LPS causes the body to initiate an innate immune response, the purpose of which is to defend the body against infection. Sepsis or septic shock is a very complex clinical condition which results when the normal host response to infection goes unchecked leading to tissue damage and ultimately multiple organ failure. Sepsis is a leading cause of death amongst critically ill patients [145]. In United States, it accounts for nearly 250,000 deaths annually [145]. The initial clinical features are characterized by fever, transient 51 hypotension, decreased urine output and thrombocytopenia which progresses into profound hypotension, coagulation abnormalities and multiple organ failure. Mononuclear cells play a key role in this process by releasing an array of pro- inflammatory cytokines and chemokines such as IL-1, IL-6, TNFa as well as other molecules such as reactive oxygen species (ROS), platelet activating factor (PAF) and nitric oxide (NO). These pro-inflammatory cytokines play an important role in sepsis by inducing a complex network of secondary responses to fight infection. 2.7.1 Animal models of sepsis There are three primary methods of inducing sepsis in animals: a) By injection of an exogeneous toxin such as LPS, b) By altering the animal’s endogenous protective barrier. This involves methods such as inducing an intestinal leakage as is done by ceacal ligation and puncture (CLP) or by colon ascendens stent peritonitis (CASP) and o) By infusing exogenous bacteria. Endotoxin (LPS) is a component of the outer membrane of gram negative bacteria and plays an essential role in the pathogenesis of sepsis. LPS administration / injection induces systemic inflammation with increase in pro-inflammatory cytokines such as TNFa and IL-1 but without bacteremia. The CLP model involves performing a surgery whereby ceacum is ligated distal to the ileocecal valve and then punctured using a needle which leads to the release of fecal contents into the peritoneum causing polymicrobial bacteremia and sepsis. The severity of sepsis produced can be adjusted based on the length of the ligated cecum as well as the size / number of punctures. The 52 bacterial infusion model of sepsis can approximate introducing a single pathogen in a controlled manner thus allowing reproducible infection. Each of these methods has its advantages and disadvantages as listed in Table 2.4. For example, LPS injection model is very simple and also a sterile method. Furthermore the dose of LPS can be titrated. This model can be used to mimic early sepsis as is seen in human patients where there is little hemodynarnic compromise. This is very useful to study systemic and renal responses during initial phases of sepsis due to the fact that a lower dose of LPS does not cause any systemic hypotension despite decreasing glomerular perfusion. CLP model has an advantage that it shows a similar cytokine profile as is seen in human sepsis. Furthermore, multiple bacterial species are observed in circulation similar to human sepsis. The disadvantage being the strain variability and also that the standard CLP model does not develop reproducible acute lung or kidney injury as seen in human sepsis. Despite the fact that sepsis is mostly characterized by the presence of multiple species of bacteria, human sepsis can also be caused by a single species. Bacterial infusion model can be used to study the sepsis caused by a single species as well as for mimicking pneumonia and other nosocomial infections. The single pathogen can be introduced in a controlled manner. This model essentially provides complimentary information in a pathogen specific manner. Out of these three different models of sepsis, LPS injection or infusion has been widely used model in sepsis research. Infusion or injection of LPS induces a systemic inflammatory response that resembles several initial clinical 53 Table 2.4: Animal models of sepsis and their advantages verses disadvantages Slightly modified from Doi, K., Leelahavanichkul, A., Yuen, PS, and Star, RA. (2009) The Journal of clinical investigation 119(10), 2868-2878 54 Animal model Advantage Disadvantage LPS injection Simple and sterile with some similarities to human sepsis pathophysiology Early and transient increase in inflammatory mediators more intense than in human sepsis Early silent period; Age and strain variability; CLP or CASP moderate and delayed peak of early hemodynarnic period in mediators; some models multiple bacterial flora Infusion or No change in intrarenal instillation of Early hyperdynamic state microcirculation; exogenous need large animals; bacteria labor intensive 55 characteristics of sepsis although there is no bacteremia. As compared to human sepsis, LPS injection/infusion however causes an earlier induction of cytokines. The cytokine levels are also higher in this case as compared to that observed in human sepsis. However, an exception to this is meningococcal sepsis as cytokine levels observed in LPS injection/infusion model are comparable to what is seen in meningococcal sepsis [146, 147]. 2.7.2 Different animal species as models for sepsis In general small animals are preferred for sepsis research due to the fact that they can be generated as genetically similar, are relatively inexpensive and can be maintained as pathogen free. However, large animals have also been utilized for this purpose. For example, pig model has been used due to the similarity of its cardiovascular, renal and gastrointestinal anatomy and physiology to that of humans. Primates, in particular baboons are immunologically similar to humans thus making them good models to study the cytokine response[l48]. There are species differences in the sensitivity to endotoxins. Moreover, within a particular species also there are differences in response based on sex, maturity levels, diet as well as estrus states. Furthermore, transgenic animal models with either deletions or overexpression of SPecific gene products are being widely used today to study the roles of particular genes. 2.7.3 Neutrophils and Monocyte/ macrophages in sepsis 56 Cells of myeloid lineage are known to play an important role in sepsis. Neutrophils are the first and most abundant cells which arrive at the infection site. These cells have large stores of proteolytic enzymes as well as have a machinery to generate reactive oxygen species (ROS) and reactive nitrogen species (RNS) which can degrade internalized pathogens. Damage to the host tissues in sepsis can also occur due to premature neutrophil activation during migration and by extracellular release of cytotoxic molecules as well as amplification of acute inflammatory responses. Neutrophils constitutively favor apoptosis which is important for resolution of inflammation and cell turnover. It is important for neutrophils to undergo apoptosis soon after they kill microbes using ROS, RNS and proteolytic enzymes since delayed clearance of neutrophils can also contribute to organ injury. Apoptotic clearance of cells induces anti-inflammatory effects in tissues. Neutrophils from patients with sepsis has been shown to have a prolonged in vitro survival as well as increased cellular activation [149]. Although molecules such as ROS and RNS have beneficial physiologic functions such as their involvement in intracellular signaling for cytokines, redox regulation as well as defense mechanisms against pathogens, overproduction or reduced scavenging of these molecules can cause oxidative / nitrosative stress which plays a key role in enhancing sepsis [150]. 2.7.4 Coagulation defect in sepsis One of the hallmark clinical features of sepsis is the disorders of coagulation. Pro-inflammatory cytokines, in particular IL-1 and IL-6 induce coagulation during sepsis. IL-10, on the other hand regulates coagulation by inhibiting the expression of 57 tissue factor (TF) on monocytes. A more severe form of coagulation is Disseminated intravascular coagulation (DIC) which is seen in 30-50% of the cases. Coagulation is initiated by the microbial components by inducing the expression of tissue factor on mononuclear and endothelial cells. The tissue factor then activates a series of proteolytic signaling cascades resulting in the formation of thrombin from prothrombin ultimately generating fibrin from fibrinogen. Furthermore, high plasma concentrations of Plasminogen-activator inhibitor type-1 (PAI-l) prevent formation of plasmin from plasminogen thereby impairing the normal regulatory fibrinolytic mechanisms (breakdown of fibrin by plasmin). Also, it has been found that a state of sepsis induces downregulation of antithrombin, protein C and tissue factor pathway inhibitor, all of which are naturally occuring anticoagulants. These proteins also possess anti-inflanunatory properties: For eg. activated protein C directly inhibits the production of TNFa by inhibiting the transcription factors NFKB and AP-l in monocytes [151]. Hence, there is an increased production and reduced removal of fibrin which causes the deposition of fibrin clots in small blood vessels, impairing tissue perfusion and ultimately resulting in an organ failure. In our sepsis studies, we observed that the blood in our myeloid specific GRK2 deleted mice starts undergoing coagulation after around 12 hours of LPS injection. Complete blood count for the blood showed no significant differences except eosinophilia in GRK2 deleted mice as compared to control mice (Discussed in chapter five). However, the importance of this observation is not clear as yet. 2.7.5 Multiple organ failure 58 The end stage in sepsis is characterized by multiple organ failure leading ultimately to death. However, the pathogenesis of multiple organ failure is highly complex and incompletely understood. Tissue hypoxia and hypoperfiision play an important role. In addition to hypoxia, dysoxia is also seen which is characterized by a state of inability to utilize the available oxygen. Furthermore, adequate oxygenation of tissues is also compromised due to the development of tissue exudates in sepsis. Microvascular occlusion occurs due to a widespread deposition of fibrin leading to tissue hypoperfusion. Cellular infiltration of tissues, in particular by neutrOphils damage tissues by releasing lysosomal enzymes as well as superoxide derived free radicals. TNFa and some other cytokines such as IL-1 causes an increased expression of inducible nitric oxide synthase (iNOS) leading to an enhanced production of nitric oxide (NO) which contributes further to vascular instability and also causes direct myocardial depression seen in sepsis [152]. Sepsis is characterized by high cardiac output and a reduced peripheral resistance due to dilatation of systemic resistance vessels which causes a progressive systemic hypotension leading finally to organ dysfunction due to impaired organ perfusion [153]. It is believed now that multiple organ dysfunction occurs due to a combination of factors such as NO overproduction, antioxidant depletion, decreased ATP concentration, and mitochondrial dysfunction [154]. 59 Hypothesis and Specific Aims GRKs, in particular GRK2 and GRK5 are highly expressed in immune cells. Furthermore, several studies have established that the expression levels of GRKs change in the immune cells such as neutrophils and macrophages under inflammatory disease conditions such as rheumatoid arthritis and sepsis. However, the physiological importance of such changes in the expression levels of GRKs is not well established. To understand the physiological importance of GRKs in immune cell signaling as well as their role in inflammatory diseases, I propose the following overall hypothesis: Hypothesis: GRKs are important regulators of inflammatory signaling in macrophages and therefore GRKs might play essential roles in the regulation of inflammatory diseases. Each of the chapters in this dissertation is dedicated to address the following specific aims: Aim 1: To determine the biochemical mechanisms by which GRKs regulate TNFa—induced signaling pathway in mouse macrophages The role of GRKs in TNFa signaling was examined using siRNA mediated knockdown as well as by overexpression techniques, using cultured macrophage cell line Aim 2: To determine the role and mechanism by which GRKs regulate inflammation in a mouse model of (_li_sease (Endotoxic shock) 60 Subaim 1: To study the role of GRK5 in inflammation in a GRK5 knockout mice using endotoxic shock model GRK5 knockout mice were procured from Jackson labs and the role of GRK5 in inflammation was investigated by comparing the levels of pro-inflammatory cytokines, survivability etc. between the knockout and wt control mice Subaim 2: To generate and characterize a myeloid cell specific GRK2 knockout mice and to study the role of GRK2 in inflammation using endotoxic shock model A GRK2 deletion specifically in myeloid cells was achieved using a Cre—loxP technology and the role of GRK2 in inflammation was investigated in these mice by comparing the levels of pro-inflammatory cytokines, survivability etc. between the knockout and littermate control mice. Importance: The findings from these studies will have a great impact on our current understanding on the roles of GRKs in inflammatory signaling. The in vivo studies in knockout mice will further strengthen our in vitro findings as well as establish a previously unidentified role of GRKs in sepsis. 61 REFERENCES Parameswaran, N., Pao, C. S., Leonhard, K. S., Kang, D. S., Kratz, M., Ley, S. C. and Benovic, J. L. (2006) Arrestin-2 and G protein-coupled receptor kinase 5 interact with NFkappaBl p105 and negatively regulate lipopolysaccharide- stimulated ERKl/Z activation in macrophages. The Journal of biological chemistry. 281, 34159-34170 Benovic, J. L., Strasser, R. H., Caron, M. G. and Lefkowitz, R. J. (1986) Beta- adrenergic receptor kinase: identification of a novel protein kinase that phosphorylates the agonist-occupied form of the receptor. Proceedings of the National Academy of Sciences of the United States of America. 83, 2797-2801 Kuhn, H. and Dreyer, W. J. (1972) Light dependent phosphorylation of rhodopsin by ATP. F EBS letters. 20, 1-6 Bownds, D., Dawes, J., Miller, J. and Stahlman, M. (1972) Phosphorylation of frog photoreceptor membranes induced by light. Nature: New biology. 237, 125-127 Sitaramayya, A. and Liebman, P. A. (1983) Phosphorylation of rhodopsin and quenching of cyclic GMP phosphodiesterase activation by ATP at weak bleaches. The Journal of biological chemistry. 258, 12106-12109 Strasser, R. H., Sibley, D. R. and Lefl 32 C5 m .E 0 1'0 2'0 30 Time (min) 94 Figure 3.1. Continued. . .. Control siRNA GRK2 siRNA 0 5 1015 0 5 10 15 (Time,min) -:‘“A- ’ “MET-P65 --- .- -- - ~ -- ---P50 ontrol siRNA GRK2 siRNA 5 10 ‘15 0 5 10 15(Time,min) I ] I—I LJ 1' NFKB binding ‘ ,1 Free probe °Io 95 Figure 3.2: Effect of different GRK2 siRNA oligos on TNFor-induced IKBa. phosphorylation , A. Raw 264.7 macrophages were transfected with either control or GRK2 specific individual oligos (siRNA GRK2 #1 (top panel) and siRNA GRK2 #2 (bottom panel) using Amaxa’s nucleofector. Forty-eight hours after transfection, cells were serum starved for ~3 hours, and stimulated or not with TNFa (25 ng/ml) for the indicated time points. Lysates were extracted and run on SDS—PAGE, transferred to nitrocellulose and then immunoblotted for plea and tubulin. Representative blots from three similar experiments are shown. B. Raw 264.7 macrophages were transfected with either control siRNA or GRK2 siRNA using Amaxa’s nucleofector. Forty-eight hours after transfection, cells were serum starved for ~3 hours, and stimulated or not with LPS (1 pg/ml) for the indicated time points. Lysates were extracted and run on SDS-PAGE, transferred to nitrocellulose and then immunoblotted with primary antibodies against IKBa and tubulin. Secondary antibodies were fluorescently labeled and the blots were developed using Odyssey’s Licor as described before [8]. Quantitation is shown as percent control. 96 ggntrol siRNA GRK2 siRNA1 O 5 15 30 0 5 15 30 (Time, min) 5...... leBd “MW Tubulin . ,.... ~ --.... ’ GRK2 Control siRNA GRK2 siRNA2 0 5 15 3O 0 5 15 30 (Time, min) .2... .. .. _. , . “- pIKBa “W-,...“fl‘rubu'in .Aagfimnih-_.g 1 ,-u 3 1.1.1,; [1. A .155”: '7 . 't ‘ _.__..__ t. . .. . . v ——- f. . .. - . . .. q—‘W ..l . . -‘ .,..- -, ,,,..——.- ”vu- ----- ..-;--—-. --‘ B. -I-Contro| siRNA -fi-GRK2 siRNA lea levels % control Time (min) 97 Figure 3.3: GRK2 over-expression enhances TNFa-induced IKBa phosphorylation in macrophages Raw264.7 macrophages were infected with adenoviruses encoding empty vector or GRK2 at 50,000 viral particles/cell. Forty-eight hours after infection, cells were semm starved for 3-4 hours, and stimulated with TNFa (25 ng/ml) for indicated times. Lysates were immunoblotted for pIKBa, IKBO. and tubulin. A representative blot is shown in (A) and quantitation in (B). *p<0.05 compared to vector. N=3. 98 Ad-Vector Ad-GRKZ 0 5 15 30 0 5 15 30 (Time,min) .. ‘g-o w“ . g M W leBa ~~~Ww Aetin ”mus—Wwwcaxz '. i .. ,M M .5»va- . . IKB“ “mama. s—;-;j.g ‘ I .1 . -_~..;l -I—Ad-Vector -A- Ad-GRK2 .3 O V 9" IKBO. phosphorylation % maximal response N 01 <11 q> 0 1'0 2'0 3'0 Time (min) 99 macrophages. Raw264.7 macrophages were transfected with adenoviruses expressing vector or GRK2 and the effect of TNFor-induced IKBa phosphorylation tested as described earlier. As predicted, over-expression of GRK2 significantly enhanced TNFor- induced IKBOL phosphorylation compared to vector controls (Fig. 3.3). Role of GRK5 in TNFor-induced IKBa phosphorylation and degradation We next tested whether other GRKs can regulate IKBa phosphorylation/degradation. We especially focused on GRK5 since we previously showed that GRK5 inhibits LPS-induced p105 phosphorylation in macrophages [8]. Interestingly, similar to the effects of GRK2, knockdown of GRK5 using siRNA pool (Fig. 3.4A) significantly inhibited TNFoc-induced IKBa phosphorylation and degradation (Fig. 3.4B, C & D). TNFa-stimulated IKBOL phosphorylation (Ser32/36) in GRK5 knockdown cells reached only 27i5% of the maximal response after 5 min compared to 100% in control cells (Fig. 3.4C). Similarly, IKBa levels after 15 min of TNFOt stimulation reached 622t6% of untreated levels in control cells but only 99:5% in GRK5 knockdown cells (Fig. 3.4D). Unlike GRK2 knockdown, basal levels of IKBOL were somewhat elevated after GRK5 knockdown (0.361i0.080 in control vs. 0.563i0.071 in GRK5 knockdown cells). Similar to GRK2 siRNAs, individual siRNAs against GRK5 also gave similar results to that of the pool (Fig. 3.5A). Interestingly, over-expression of GRK5 using adenovirus only modestly enhanced IKBOt phosphorylation (Fig. 3.58). Role of GRKs is specific for IKBa-NFKB pathway 100 Figure 3.4: GRK5 knockdown inhibits TNFoi-induced IKBa phosphorylation and degradation in macrophages Raw 264.7 macrophages were electroporated with either control or GRK5 siRNA as indicated. Forty-eight hours after electroporation, cells were serum starved for ~3 hours, and stimulated with TNFa (25 ng/ml) for the indicated times. Lysates were immunoblotted for pIKBa, IKBO. and tubulin and blots developed using Licor’s Odyssey. Representative blots for GRK5 knockdown is shown in (A), pIKBa and IKBOt in (B). Quantitation for IKBOt phosphorylation and IKBOL degradation are shown in (C) and (D) respectively. ***p<0.001 compared to control. N=6. 101 Con GRK5si Control siRNA GRK5 siRNA 0 5 15.30 0 5 15 30(Time,min) . .14 ~ ~M -W . 9......» t... w leBa “w--~-—.—'TUPUIIH 102 Figure 3.4. Continued.... -l- Control siRNA -A— GRK5 siRNA 100- on ‘P o: i A O 1 lea phosphorylation % maximal response N 9 O 1'0 21) 30 Time (min) 0- D_ -I-Contro| siRNA --A- GRK5 siRNA 105-1 (0 O n N 01 l O) N (Di—1r 9 lea levels % control Time (min) 103 Figure 3.5 Effect of a different GRK5 siRNA oligo on TNFa—induced IKBa phosphorylation A. Raw 264.7 macrophages were transfected with either control or GRK5 specific individual oligo (siRNA GRK5 #1) using Amaxa’s nucleofector. Forty-eight hours after transfection, cells were serum starved for ~3 hours, and stimulated or not with TNFa (25 ng/ml) for the indicated time points. Lysates were extracted and run on SDS-PAGE, transferred to nitrocellulose and then immunoblotted for pIKBa and tubulin. Representative blots from three similar experiments are shown. B. Raw264.7 macrophages were infected with adenoviruses encoding empty vector or GRK5 (kindly provided by Dr. W. Koch, Thomas Jefferson University) at 1000 viral particles/cell. Forty-eight hours after infection, cells were serum starved for 3-4 hours, and stimulated with TNFa (25 ng/ml) for indicated times. Lysates were immunoblotted for pIKBa, and tubulin. A representative blot from two such experiments is shown. 104 Control siRNA GRK5 siRNA1 0 5 15 30 0 5 1.5. 30(Time,min) a... (pIKBG —.____._...__. _._::_".__~ -—-:-3 m" '7' ' Tubulin \ .. -..-.... ...-., T\ ”Q :7... :fi “'7‘ PM“ MGRK5 Ad-Vector Ad'GRKS 0 515 30 0 5 15 30 (Time,min) pled 105 In addition to IKBa phosphorylation, TNFor treatment also induces NFKBl p105 (another member of the IKB family) phosphorylation at Ser932 [28]. Previous studies have shown that GRK2 and 5 can interact with p105 and that GRK5 negatively regulates LPS- induced p105 phosphorylation [8]. To determine whether the observed effects of GRK2/5 knockdown are specific for TNFoc-induced IKBOt phosphorylation or whether p105 phosphorylation can also be regulated in a similar manner, we examined TNFor-induced p105 phosphorylation in control and GRK2/5 knockdown macrophages. Interestingly, unlike IKBa phosphorylation, GRK2/5 knockdown did not affect TNFor—induced p105 phosphorylation (at Ser932) (Fig. 3.6A and 3.6B). This suggests that the role of GRK2/5 is specific for TNFor-induced IKBa-NFKB pathway. Regulation of NFKB-dependent gene expression by GRK2 and Sin macrophages IKBa-NFKB pathway was recently shown to be critical for the expression of MIPlB (macrophage inflammatory protein 1B), one of the major chemokines expressed by macrophages [29]. Therefore, we tested whether the role of GRK2 and 5 on the TNFor-induced NFKB pathway extended downstream of NFKB activation to MIPlB mRNA expression. Treatment of control macrophages with TNFoc induced MIPlB mRNA expression by ~4-5-fold. This increase in MIPlB expression was significantly blocked in both GRK2 and 5 knockdown macrophages (Fig. 3.7A and 3.7B). These results demonstrate that GRK2 and 5 regulate TNFor-induced IKBOi-NFKB pathway as well as its physiological gene expression target in macrophages. Kinase activity of GRK2 and 5 is required for TNFa-induced NFKB-dependent gene transcription 106 Figure 3.6 Role of GRKs is specific for IKBa-NFKB pathway A. Raw 264.7 macrophages were transfected with either control siRNA or GRK2 siRNA using Amaxa’s nucleofector. Forty-eight hours after transfection, cells were serum starved for ~3 hours, and stimulated or not with TNF or (25 ng/ml) for the indicated time points. Lysates were extracted and run on SDS-PAGE, transferred to nitrocellulose and then immunoblotted with primary antibody against phospho-p105. For quantitation, p-p105 bands were normalized as described before [8]. Quantitation is shown as percent maximal response. N=4. B. Raw 264.7 macrophages were transfected with either control siRNA or GRK5 siRNA using Amaxa’s nucleofector and treated as described above in A. Quantitation is shown as percent maximal response. N=4. 107 10 20 30 Time (min) -A- GRK2 siRNA 0 1251 -I—Control siRNA 125' -I—Contro| siRNA owwoamo. .mE_me xx. cozflbocamofi morn A. d 5 7 omMoawo. .2:me xx. cougbocamoca moE O 5 10 2'0 30 Time (min) 108 Figure 3.7 Expression of Macrophage inflammatory protein-1B (MIPl B), an NFKB-regulated gene, is inhibited by GRK2 or GRK5 knockdown in macrophages A. Control or GRK2 knockdown Raw264.7 macrophages were treated or not with TNFor for 24 hours and RNA extracted as described in the Methods. Quantitative real-time RT-PCR was performed using MIPIB specific primers. MIPlB mRNA levels were normalized to cyclophilin levels. ** p<0.01. N=3. B. Control or GRK5 knockdown Raw264.7 macrophages were treated with TNFor and mRNA levels of MIPl B and cyclophilin determined as described in (A) above. *** p<0.001; **p<0.01. N=3. 109 EControl siRNA -GRK2 siRNA **, [— A O O J MIP1B mRNA % maximal response 01 \I ‘2 3‘ N 01 l 0 24 Time (hr) 0 L DControl siRNA RK ' *** -G 5 SIRNA _ 1001 —* G) (D 5 75 En ‘ 0:8 5% 0:. ti. (U 2's o\° Time (hr) 110 Figure 3.8 Kinase activity of GRK2 and 5 is essential for mediating TNFa-induced NFKB transcriptional activation A. HEK293T cells were transfected with vector, GRK2 wild type or GRK2- K220R (kinase deficient mutant) expression plasmids along with pELAM- luciferase and LacZ. Forty hours after transfection, cells were stimulated (in triplicates) with different concentrations of TNFa (as indicated) for 8 hours. Lysates were prepared and analysed for luciferase and B-galactosidase activities. Quantitation was performed after normalizing luciferase activity to B-galactosidase activity. Data is expressed as fold basal of the vector transfected cells. Immunoblots showing expression levels of GRK2 wild type and GRK2 kinase deficient mutant are shown in the top panel. *P<0.05. N=4. B. HEK293T cells were transfected with vector, GRK5 wild type or GRK5- K215R (kinase deficient mutant) expression plasmids along with pELAM- luciferase and LacZ. Cell treatments, activity assays and quantitation were performed as described in (A) above. Irnmunoblots showing expression levels of GRK5 wild type and GRK5 kinase deficient mutant are shown in the top panel. **P<0.0l N=4. 111 NFKB-luciferase activity (Fold basal) NFKB-lucrferase actrvrty (Fold basal) 8 K3 6‘. C? ‘1‘ ‘9 WT KinD ~ ~ GRK2 250' EVector 225. -GRK2 WIId type MGRKZ kinase dead 200- 1 75- V 01 I N 01 (II 0 l I 0‘ 0 5 TNFa(ng/ml) WT KinD -~GRK5 250- 1:1Vector 225- -GRKS wild type 200_ 1;; GRK5 kinase dead 175- 15m 1.. 125- 100- 75- 50- 25- 0_ ; 0 5 10 25 TNFa(ng/ml) 112 To further explore the biochemical mechanism by which GRK2 and 5 mediate TNFor-induced NFKB activity, we next tested whether the kinase activity is essential for the observed effects of the GRKs. For this, we over-expressed vector or wild type or kinase dead GRK2 or GRK5 in HEK293T cells along with an NFKB reporter plasmid (pELAM luciferase) and LacZ (for transfection normalization). Forty-hours after transfection, cells were serum starved (~3 hours) and were stimulated (or not) with TNFor for 8 hours and luciferase and B-galactosidase activity determined as described in the methods. In vector transfected cells, TNFa stimulation significantly increased NFKB- luciferase activity (Fig. 3.8). As predicted, over-expression of wild type GRK2 or GRK5 significantly enhanced TNFa-induced NFKB activity while over-expression of kinase- inactive GRK2 or GRK5 (GRK2-K220R or GRK5-K215R) had no effect (Fig. 3.8A and 3.8B). This demonstrates that the kinase activities of GRK2 and 5 are required for the observed effects of GRKs in TNFor-induced NFKB signaling. Interestingly, as observed with IKBa phosphorylation in macrophages, over-expression of GRK2 but not GRK2- K220R enhanced NFKB-luciferase activity even in the absence of ligand stimulation (1.0 i 0.51 activity in vector cells compared to 2.35 at 1.07 in GRK2 and 0.70 :t 0.32 in GRK2-K220R expressing cells). In contrast, over-expression of GRK5 or GRK5—K215R did not significantly affect basal NFKB activity (1 i 0.14 activity in vector cells compared to 1.51 :t 0.17 in GRK5 and 0.86 i 0.04 in GRK5-K215R expressing cells). Interaction of GRK2 and 5 with NBC: To define the biochemical mechanisms that mediate GRKs’ actions, we first tested whether GRKs affect H(KB expression or activity. For this purpose, we knocked down and over-expressed GRK2/5 in Raw264.7 macrophages and HEK293T cells 113 respectively and examined the levels of IKKB. Neither knockdown nor over-expression of GRK2/5 affected IKKB levels in the presence or absence of ma (data not shown). To further rule out the effect of GRKs on H(KB activity, we performed an in vitro IKKB kinase assay using IKKB immunoprecipitated from cells over-expressing GRK2/5. In vitro phosphorylation of GST-IKBOL by immunoprecipitated IKKB was not affected by over-expression of either GRK2 or GRK5 (data not shown). Similarly, the interaction of IKBor and IKKB was not affected by over-expression of GRKs in HEK293T cells (data not shown). Based on these results we hypothesized that GRKs might mediate their effects via directly interacting with and phosphorylating IKBa. To first examine if GRK2 or 5 can interact directly with IKBOL, we tested the ability of GST or GST-IKBa to bind GRK2 and 5 using an overlay blot assay. For this, bacterially expressed and purified GST or GST-IKBOL or GST-IKBa(76-302) were run on SDS-PAGE, and transferred to nitrocellulose. The membranes were then incubated with HEK293T lysates over- expressing either vector, GRK2 or GRK5 and tested for the ability of GRKs to specifically bind to GST-IKBOL. As hypothesized, both GRK2 (Fig. 3.9A and 3.9B) and GRK5 (Fig. 3.10A and 3.10B) bound to GST-IKBOL with no significant binding to GST. In addition, neither GRK2 (Fig. 3.9A) nor GRK5 (Fig. 3.10A) interacted appreciably with an N-terminal deletion mutant of IKBa [IKBa(76-302)], suggesting that both GRKs primarily interact with IKBa at the N-terminus. We also tested the ability of GRKs to interact with IKBa in intact cells and found that immunoprecipitation of IKBa co- immunoprecipitated both GRK2 and 5 (Fig. 3.9C and 3.10C) in HEK293T cells; Fig. 3.9D in human monocytic cells THPl). Taken together, these results suggest that IKBCX is 114 Figure 3.9 GRK2 interacts with the N-terminus of IKBa A. Purified GST-IKBa (full length), GST-IKBa (76-302) and GST alone, (5 pg each) were resolved on a SDS-PAGE and transferred to a nitrocellulose membrane. The membrane was incubated overnight in HEK293T lysates over-expressing HA-GRK2 (A) or vector (B). Membrane was washed and probed for HA-GRK2 binding to GST-IKBOL by immunoblotting using anti- HA antibody. Western blot of the overlay assay is shown in the top panel and a Ponceau stain of the overlay blot is shown in the bottom. Blots are representative of at least four individual experiments. C. GRK2 and IKBa were co-expressed in HEK293T cells and the lysates were immunoprecipitated using IKBa polyclonal antibody and the blots were then subjected to immunoblotting as indicated. D. Lysates from THPl monocytic cells were immunoprecipitated with IKBa polyclonal antibody. Immunoprecipitates were then denatured using sample buffer, boiled and separated on a 10% SDS-PAGE gel. Irnmunoblotting was performed for GRK2 as shown. 115 Overlay: HAGRK2 Overlay: Vector WBzHA WBzHA s" , )s \ 4‘ 9° 9" 0” 9° 9 + :5 09" 09") (\Q’ 09" ’9‘ 0 ’\ I _Total Protein . Total Protein un- . m % GRK2 + + IKBa + + IKBa peptide + - _ "' IB GRK2 IPIKBo ' 71' IBIKBa - IPIKBa D . Antl-IKBOL CfllgG 106 m 13 GRK2 IPIKBa { . _ IBIKBd - ..._... “h- IPIxBa 116 Figure 3.10 Direct interaction between GRK5 and IKBa A. Purified GST-IKBa (full length), GST-IKBa (76-302) and GST alone, (5 pg each) were resolved on a SDS-PAGE and transferred to a nitrocellulose membrane. The membrane was incubated overnight in HEK293T lysates over-expressing GRK5 (A) or vector (B). Membrane was washed and probed for GRK5 binding to GST-IKBOL by immunoblotting using anti-GRK5 antibody. Western blot of the overlay assay is shown in the top panel and a Ponceau stain of the overlay blot is shown in the bottom. Blots are representative of at least four individual experiments. C. GRK5 and IKBa were co-expressed in HEK293T cells and the lysates were immunoprecipitated using IKBa polyclonal antibody and the blots were then subjected to immunoblotting as indicated. 117 Overlay: GRK5 WB: GRK5 0 543’ « C96" 96‘ \ .g' 9 :5 OQQ, 96 Overlay: Vector WB: GRK5 o 0 .1. 9° Ix" a) Q 4 C9 GRK5 IKBa IKBci peptide _, + + T" , lB GRK5 . IPIKBa 4. 18 “(Ba IP IKBu 118 a direct interaction partner for both GRK2 and GRK5. Phosphorylation of IKBa by GRK2 and 5 Experiments using kinase-dead mutants of GRK2 and 5, as well as the experiments described above, suggest that IKBa may be a substrate for GRKs in TNFa-induced NF KB signaling. To directly test this, we performed in vitro phosphorylation assays using purified GRK2 and 5 with IKBa as the substrate. GRK5 effectively phosphorylated IKBa to a stoichiometry of ~0.75 mol/mol (Fig. 3.11B) and interestingly, phosphorylation was effectively attenuated by the addition of phospholipids, which normally activate GRK5 (Fig. 3.11D) [30]. In contrast, IKBa was a relatively poor substrate for GRK2 (Fig. 3.11A), although the phosphorylation was enhanced by the addition of GBy subunits and phospholipids, known activators of GRK2 (Fig. 3.11C) [31]. Taken together, these results demonstrate that IKBa is an in vitro substrate for both GRK2 and GRK5. To identify the GRK phosphorylation sites in IKBct and to examine if IKBa is differentially phosphorylated by GRK2 and 5, we first tested whether the known IKKB phosphorylation sites (Ser32/36) are also phosphorylated by GRK2 or GRK5. Indeed, previous studies have shown that these two residues are targeted by additional kinases such as ribosomal S6K and CK 11, especially in NFKB pathways that are largely IKKB- independent [32, 33]. To test this, we assessed the ability of GRK2 and GRK5 to phosphorylate a GST-IKBa S32/36A mutant. Our results show that GRK2 mediated phosphorylation of wild type and mutant GST-IKBoi was comparable, suggesting that these sites are not phosphorylated by GRK2 (Fig. 3.11E). In contrast, GRK5 mediated phosphorylation of the IKBO. mutant was decreased ~60% compared to wild type IKBa 119 Figure 3.11 IKBa is a substrate for GRK2 and GRK5 in vitro A and B. Purified GRK2 and GRK5 were prepared as described before [30]. 200 nM purified GST-IKBa or GST alone were incubated with 25 nM GRK2 or GRK5 in buffer containing 20 mM Tris-HCl, pH 7.5, 2 mM EDTA, 0.1 mM [y32P] ATP, 5 mM MgC12 and the phosphorylation reactions were done for various time points as indicated. Reaction was stopped with SDS buffer, electrophoresed on a 10% SDS-PAGE, the gel dried and subjected to autoradiography. Phosphorylated IKBd bands were then counted on a liquid scintillation for quantification. C and D. The reactions were incubated in the absence or presence of liposomes as indicated and phosphorylation reactions performed for 30 min and stoichiometry quantified as described above. GRK2 phosphorylation samples also contained GBy in addition to liposomes. E and F. GRK2 and GRK5 were incubated with 200 nM purified GST-IKBa (wild type or mutant S32A/S36A) in a kinase reaction and phosphorylated IKBa quantified using liquid scintillation as described in the methods. G. Purified GRK2, GRK5 or IKKB were incubated with GST-IKBa as a substrate as described above without [y3zP]ATP. Reaction was stopped with SDS buffer, electrophoresed on a 10% SDS-PAGE and transferred to a nitrocellulose membrane. Immunoblotting was performed using an antibody that specifically detects phospho-IKBOL(ser32). 120 mol Pi/mol IKBO. mol Pi/mol IKBa GRK2 0.125- 0.100- 0.075- 0.05% 0.025- 0.000d 0.12- 0.101 0.08-1 0.06- 00+ 0.02- o 5 1'0 1'5 2'0 2'5 3'0 Time (min) 0.00 GRK2 GRKZ‘I'LIp‘I'GBy 121 mol Pi/mol lkBa U mol P1/mol IKBa 9.0.0 #016) JII GRK5 .9 .o ." 01 N O O 01 O I 4 I 0.3-1 9 N I 0.11 5 1'0 1'5 2'0 2530 Time (min) GR'K5 GRK5 + Lip Figure 3.1 1 Continued... 0.14- 0.12- 0.10- 0.081 0.06- 0.04- 0.021 mol Pi / mol IKBOl. DIKBOI WT - IKBOL (S32A, S36A) __|_ 0.00 0.6- m 0.5- : 0.4- E 0.3- E 0.2- E (11. 0.0 GRK2 I:I IKBo. WT - IKBOL ($32A, S36A) T 122 Figure 3.11 Continued... W ‘0 Kinase g f $2 I:I GST-lKBOi .GST-IKBOL + GRK2 {>3 .GST-IKBor + GRK5 P .GST-IKBOt + IKKB a: 100- L”, E: 71? 75- E g e . €012 50 8 i 25- .C Q. 58 0' a 123 (Fig. 3.11F), suggesting that GRK5 phosphorylates one or both of these sites. This result was confirmed using an antibody that recognizes phosphoSer32 in IKBa. For these studies, GST-IKBa was initially phosphorylated in vitro using GRK2, GRK5 or IKKB and the samples were run on SDS PAGE and immunoblotted using anti-IKBa-phospho-Ser32. These results show that Ser32 is selectively phosphorylated by GRK5 but not GRK2 and that GRK5 mediated phosphorylation of IKBa is comparable to that seen with IKKB (Fig. 3.11G). Overall, these results reveal that Ser32 is phosphorylated by GRK5 and that GRK2 and GRK5 phosphorylate distinct residues. IKKB knockdown does not inhibit TNFor-induced IKBa phosphorylation in Raw264.7 macrophages Similar to GRK5, other kinases such as casein Kinase II and ribosomal S6 kinase have been shown to phosphorylate IKBa at Ser32/36. Interestingly, these kinases were shown to selectively regulate IKBOt-NFKB pathway in an IKKB-independent manner. Therefore, we hypothesized that because of the role of GRK2 and 5 as “IKBa kinases”, IKKB may be dispensable in Raw264.7 cells, particularly for TNFa-induced IKBa phosphorylation. To test this hypothesis, we knocked down IKKB in Raw264.7 macrophages and tested the effect of TNFor on IKBOL phosphorylation (Ser32/36). As predicted, we found that TNFor-induced IKBa phosphorylation is not significantly affected by knockdown of IKKB, suggesting that H(KB may be redundant in this system (Fig. 3.12A). However, it is possible that the level of knockdown is not sufficient to inhibit IKBa phosphorylation because of the residual IKKB kinase activity present. To rule out this possibility, we further tested the ability of IKKB knockdown to inhibit LPS- induced IKBa phosphorylation. Interestingly, our results demonstrate that LPS-induced 124 Figure 3.12 IKKB knockdown does not inhibit TNFa-induced IKBa phosphorylation but inhibits LPS-induced phosphorylation A. Raw 264.7 macrophages were electroporated with either control or IKKB siRNA as indicated. Forty-eight hours after electroporation, cells were serum starved for ~3 hours, and stimulated with TNFa (25 ng/ml) for the indicated times. Lysates were innnunoblotted for pIKBa (Ser32/36) and tubulin for normalization. Representative blots are shown at the top and quantitation at the bottom. N=4. B. Raw 264.7 macrophages were electroporated with either control or IKKB siRNA as indicated. Forty-eight hours after electroporation, cells were serum starved for ~3 hours, and stimulated with LPS (1 pg/ml) for the indicated times. Lysates were immunoblotted for pIKBa (Ser32/36) and tubulin for normalization. Quantitation is shown. N=4. 125 Control siRNA IKKB siRNA TNFa 0 5 15 30 0 515 30 Time(min) .. .1. w ._.. —. “mm, ~ , M" leBa IKKB .2...— Tubulin wwwwwvmw.—.i-v ‘°°' DControl siRNA g 3,? % -lKKbeta siRNA ’- = 75- 2 a E‘ I!) g 2 Q'- 50- M £5 a 5 :5 E 25- % .\° 0' h 0 5 15 30 Time (min) DControl siRNA .lKK-beta siRNA 100- : A o a 7 I “5 5 _ Q. 75- E‘ m g 2 Q— a § _§ 50- * O- 5 , 33 E 25- 5 o\ V 1 c . .J 0 1o 30 60 Time (min) 126 IKBa phosphorylation (at Ser32/36) is inhibited by IKKB knockdown and this effect was particularly evident at later time points (Fig. 3.12B). These results suggest that the IKKB plays a crucial role in LPS-induced IKBa phosphorylation, but not in TNFor-induced IKBOi-NFKB pathway in Raw264.7macrophages. Taken together, our results demonstrate a critical role for GRK2 and 5 in the regulation of TNFor-induced IKBOt—NFKB pathway in Raw264.7 macrOphages and suggest that IKBa phosphorylation by GRKs might be an essential step in this regulation. 127 DISCUSSION G-protein coupled receptor kinases were first discovered for their role in GPCR phosphorylation and desensitization [34, 35]. Recent studies, however, have revealed a number of non-GPCR substrates for GRKs [10, 11]. Although GRKs mediate their cellular effects for the most part through their catalytic activity, recent studies have also proposed a kinase-independent role for GRKs in cellular signaling (via protein-protein interaction with the RH domain). In this regard, GRK2 has been shown to interact with MEKl and regulate ERK activation in a kinase-independent manner [36]. GRK2 has also been shown to interact with other proteins such as PI3K [3 7], Akt [3 8], and GIT [39] and regulate a number of cell biological effects. In addition to the receptor and cytosolic substrates, GRKs, have also been found to phosphorylate nuclear proteins. A physiologically important role for the nuclear localized GRK5 was recently identified by Martini et al [40] who showed that the nuclear GRK5 is a HDAC kinase the mediates the epigenetic regulation of gene expression in cardiomyocytes. Taken together these studies suggest that the role of GRKs is much broader than previously appreciated. Studies have just begun to emerge on the potential role for GRKs in the regulation of various components of the NFKB pathway. In this regard, we demonstrated that GRK5 stabilizes LPS-stimulated p105 levels in macrophages [8]. More recently, while this manuscript was in preparation, a similar stabilizing role for GRK5 in maintaining IKBOL levels in endothelial cells was shown [41]. Surprisingly in the present study we find that GRK2 and GRK5 are important regulators of IKBa-NFKB signaling and mediate TNFa- induced IKBa phosphorylation. In addition, in contrast to the findings of Sorriento et al [41] in endothelial cells, our results demonstrate that GRK2 and GRK5 mediate TNFor- 128 induced NF KB-dependent gene transcription in macrophages. Our results further suggests that even with in a given cell type, the role of GRKs is selective for a particular ligand. Our data further demonstrate that the role of GRK2 and GRK5 on TNFa-induced NFKB signaling is dependent on the kinase activities because the kinase-deficient GRKs failed to mediate NFKB activation. Also, in vitro kinase assays indicate that IKBOL may be differentially phosphorylated by GRK2 and 5. Our results show that the presence of GBy subunits and liposomes significantly enhances the phosphorylation of IKBOt by GRK2. GBy subunits have clearly been demonstrated to be important in the translocation of GRK2 to the plasma membrane for GPCR phosphorylation. Whether a similar role for GBy subunits in TNFa signaling exists, is presently not known. However, Kawamata et al [42] showed recently that TNFor signaling is mediated by activation of G-proteins in adipocytes. In addition, TNFor treatment of THP-l monocytic cells has been shown to mediate GRK2 translocation to the membrane and affect B-adrenergic receptor desensitization [43], suggesting possible regulation of TNFor-induced GRK2 activity by G-proteins. In contrast to the role of lipids in GRK2 activity, GRK5 phosphorylation of IKBa appears to be effectively inhibited in the presence of lipids even though previous studies have clearly shown that lipids activate GRK5 [30]. Thus it is possible that GRK5 phosphorylation of IKBOt is regulated by biochemical mechanisms that are distinct from its phosphorylation of other substrates such as synucleins [30] as well as from that of IKBa phosphorylation by GRK2. Whether these differences in the phosphorylation of IKBa by GRK2 and 5 translate into regulation of IKBOL in different sub-cellular enviromnents in not known and will be tested in future studies. Other studies have clearly 129 shown that IKBa-NFKB complexes can be present in different sub-cellular environments [44, 45] and therefore, these complexes could be potentially regulated by GRK2 or 5 depending on the local cellular environment. IKKB has been identified as the primary kinase that phosphorylates IKBOL. However, there is now extensive evidence that other kinases including IKKa, CK II and ribosomal S6K can phosphorylate IKBO. at the same sites as that of IKKB. This redundancy can in part be explained by the receptor- and cell type-specific regulation of IKBor-NFKB pathways [7, 32, 46]. For example, studies have shown that UV light- induced IKBOL degradation is mediated by phosphorylation of IKBoc by CK II [7]. Also, PMA-induced IKBor phosphorylation has been shown to involve ribosomal S6K [32]. Similarly, recent studies have shown IKKB-dependent and —independent pathways that regulate IKBOL-NFKB pathway in human macrophages in response to specific ligands [47]. Our studies clearly suggest that GRKs while necessary for TNFor-induced IKBa- NFKB pathway, are not involved in LPS-induced IKBa-NFKB signaling. Interestingly, knockdown of IKKB does not affect TNFoc-induced IKBa phosphorylation (at Ser32/36), but does inhibit LPS-induced IKBOL phosphorylation, suggesting that in Raw264.7 macrophages, GRKs play the role of IKBa kinases selectively for TNFoc signaling. Our in vitro kinase reactions further support our findings in macrophages in that, GRK5 is able to phosphorylate some residues in IKBOL that are similar to that of IKKB. Thus it is possible that GRK5 specifically might function in a similar capacity to that of IKKB. If GRK5 can function as an IKBOL kinase, then what is the role of GRK2? It appears from our macrophage experiments that GRK2 is also necessary for TNFor-induced IKBOL 130 phosphorylation (Ser32/36). However, in vitro GRK2 does not phosphorylate Ser32/36 and therefore appears to phosphorylate a different set of residues. In cells, GRK2 phosphorylation of these unidentified residues appears to be necessary for phosphorylation of Ser32/36 since knockdown of GRK2 inhibits IKBa phosphorylation at these two sites. It is also possible the level of IKKB knockdown obtained might not be sufficient to define IKKB-independent regulation because of the residual kinase activity present in the knockdown cells. If this is the case, GRKs might work co-operatively with IKKB in the phosphorylation of IKBa. In this regard, although Ser32/36 in IKBOt are the well-characterized IKK phosphorylation sites, there is strong evidence that IKK also phosphorylates less well-characterized sites at the c-tenninus [48]. Therefore, in our experiments, we carmot rule out the role of IKKB on phosphorylation of these other sites. Our results, however, indicate that H(KB knockdown can certainly inhibit LPS-induced IKBa phosphorylation (on Ser32/36) and therefore, suggest that LPS and TNFa signal to NFKB activation via different mechanisms in Raw264.7 macrophages. In conclusion, our studies unravel important biochemical roles for GRK2 and 5 in TNFor-induced IKBOL phosphorylation and NFKB signaling. Because regulation of the NFKB pathway appears to be receptor-specific as well as cell type-specific, further broad and unbiased proteomic approaches are necessary to identify macrophage-specific and TNFR-specific signaling complexes that mediate NFKB activation. These studies will undoubtedly identify therapeutic targets for inhibiting TNFa signaling in chronic inflammatory diseases. 131 Acknowledgement: We would like to thank Dr. Christina Pao and Ms. Michelle Kratz for preparing purified GRKs. This work was funded in part by grants from National Institutes of Health AR055726 and HL095637 (to NP.) and GM44944 (to J LB.) and grant-in-aid from the Midwest affiliate of American Heart Association (to NR). 132 REFERENCES Le, J. and Vilcek, J. (1987) Tumor necrosis factor and interleukin 1: cytokines with multiple overlapping biological activities. Laboratory investigation; a journal of technical methods and pathology. 56, 234-248 Reich, K., Nestle, F. 0., Papp, K., Ortonne, J. P., Evans, R., Guzzo, C., Li, S., Dooley, L. T. and Griffiths, C. E. (2005) Infliximab induction and maintenance therapy for moderate-to-severe psoriasis: a phase III, multicentre, double-blind trial. Lancet. 366, 1367-1374 Song, H. Y., Regnier, C. H., Kirschning, C. J., Goeddel, D. V. and Rothe, M. (1997) Tumor necrosis factor (TNF)-mediated kinase cascades: bifiircation of nuclear factor-kappaB and c-jun N-terminal kinase (JNK/SAPK) pathways at TNF receptor-associated factor 2. Proceedings of the National Academy of Sciences of the United States of America. 94, 9792-9796 Baldwin, A. 8., Jr. (2001) Series introduction: the transcription factor NF- kappaB and human disease. The Journal of clinical investigation. 107, 3-6 Bonizzi, G. and Karin, M. (2004) The two NF-kappaB activation pathways and their role in innate and adaptive immunity. Trends in immunology. 25, 280- 288 Basak, S., Kim, H., Keams, J. D., Tergaonkar, V., O'Dea, E., Werner, S. 1..., Benedict, C. A., Ware, C. F., Ghosh, G., Verma, I. M. and Hoffinann, A. (2007) A fourth IkappaB protein within the NF-kappaB signaling module. Cell. 128, 369-381 Kato, T., Jr., Delhase, M., Hoffinann, A. and Karin, M. (2003) CK2 Is a C- Terminal IkappaB Kinase Responsible for NF-kappaB Activation during the UV Response. Molecular cell. 12, 829-839 Parameswaran, N., Pao, C. S., Leonhard, K. S., Kang, D. S., Kratz, M., Ley, S. C. and Benovic, J. L. (2006) Arrestin-2 and G protein-coupled receptor kinase 5 interact with NFkappaBl p105 and negatively regulate lipopolysaccharide- stimulated ERK1/2 activation in macrophages. The Journal of biological chemistry. 28], 34159-34170 133 10 ll 12 13 14 15 16 17 Premont, R. T. and Gainetdinov, R. R. (2007) Physiological roles of G protein- coupled receptor kinases and arrestins. Annual review of physiology. 69, 511- 534 Cant, S. H. and Pitcher, J. A. (2005) G protein-coupled receptor kinase 2- mediated phosphorylation of ezrin is required for G protein-coupled receptor- dependent reorganization of the actin cytoskeleton. Molecular biology of the cell. 16, 3088-3099 Freeman, J. L., De La Cruz, E. M., Pollard, T. D., Lefkowitz, R. J. and Pitcher, J. A. (1998) Regulation of G protein-coupled receptor kinase 5 (GRK5) by actin. The Journal of biological chemistry. 273, 20653-20657 Wu, J. H., Goswami, R., Cai, X., Exum, S. T., Huang, X., Zhang, L., Brian, L., Premont, R. T., Peppel, K. and Freedman, N. J. (2006) Regulation of the platelet-derived growth factor receptor-beta by G protein-coupled receptor kinase-5 in vascular smooth muscle cells involves the phosphatase Shp2. The Journal of biological chemistry. 28], 37758-37772 Kong, G., Penn, R. and Benovic, J. L. (1994) A beta-adrenergic receptor kinase dominant negative mutant attenuates desensitization of the beta 2- adrenergic receptor. The Journal of biological chemistry. 269, 13084-13087 Levay, K., Satpaev, D. K., Pronin, A. N., Benovic, J. L. and Slepak, V. Z. (1998) Localization of the sites for Ca2+-binding proteins on G protein- coupled receptor kinases. Biochemistry. 37, 13650-13659 Van Antwerp, D. J., Martin, S. J., Kafri, T., Green, D. R. and Verma, I. M. (1996) Suppression of TNF-alpha-induced apoptosis by NF-kappaB. Science (New York, NY. 274, 787-789 Nakano, H., Shindo, M., Sakon, S., Nishinaka, S., Mihara, M., Yagita, H. and Okumura, K. (1998) Differential regulation of IkappaB kinase alpha and beta by two upstream kinases, NF-kappaB-inducing kinase and mitogen-activated protein kinase/ERK kinase kinase-1. Proceedings of the National Academy of Sciences of the United States of America. 95, 3537-3542 Wu, C. J., Conze, D. B., Li, T., Srinivasula, S. M. and Ashwell, J. D. (2006) Sensing of Lys 63-linked polyubiquitination by NEMO is a key event in NF- kappaB activation [corrected]. Nature cell biology. 8, 398-406 134 l8 19 20 21 22 23 24 25 26 Gao, H., Sun, Y., Wu, Y., Luan, B., Wang, Y., Qu, B. and Pei, G. (2004) Identification of beta-arrestin2 as a G protein-coupled receptor-stimulated regulator of NF-kappaB pathways. Molecular cell. 14, 303-317 Latz, E., Verma, A., Visintin, A., Gong, M., Sirois, C. M., Klein, D. C., Monks, B. G., McKnight, C. J ., Lamphier, M. S., Duprex, W. P., Espevik, T. and Golenbock, D. T. (2007) Ligand-induced conformational changes allosterically activate Toll-like receptor 9. Nature immunology. 8, 772-779 Schindler, U. and Baichwal, V. R. (1994) Three NF-kappa B binding sites in the human E—selectin gene required for maximal tumor necrosis factor alpha- induced expression. Molecular and cellular biology. 14, 5820-5 831 Bhattacharyya, R. and Wedegaertner, P. B. (2003) Characterization of G alpha lB-dependent plasma membrane recruitment of p115RhoGEF. The Biochemical journal. 37], 709-720 Loniewski, K., Shi, Y., Pestka, J. and Parameswaran, N. (2008) Toll-like receptors differentially regulate GPCR kinases and arrestins in primary macrophages. Molecular immunology. 45, 2312-2322 Li, J., Joo, S. H. and Tsai, M. D. (2003) An NF-kappaB-specific inhibitor, IkappaBalpha, binds to and inhibits cyclin-dependent kinase 4. Biochemistry. 42, 13476-13483 Bomberger, J. M., Spielman, W. S., Hall, C. S., Weinman, E. J. and Parameswaran, N. (2005) Receptor activity-modifying protein (RAMP) isoforrn-specific regulation of adrenomedullin receptor trafficking by NHERF- ]. The Journal of biological chemistry. 280, 23926-23935 Chuang, T. T., Sallese, M., Ambrosini, G., Parruti, G. and De Blasi, A. (1992) High expression of beta-adrenergic receptor kinase in human peripheral blood leukocytes. Isoproterenol and platelet activating factor can induce kinase translocation. The Journal of biological chemistry. 267, 6886-6892 London, R. P., Perussia, B. and Benovic, J. L. (1996) Differentially regulated expression of the G-protein-coupled receptor kinases, betaARK and GRK6, during myelomonocytic cell development in vitro. Blood. 88, 4547-4557 135 27 28 29 3O 31 32 33 34 35 Dicharnp, 1., Bourgeois, A., Dirand, C., Herbein, G. and Wendling, D. (2007) Increased nuclear factor-kappaB activation in peripheral blood monocytes of patients with rheumatoid arthritis is mediated primarily by tumor necrosis factor-alpha. The Journal of rheumatology. 34, 1976-1983 Lang, V., Janzen, J., Fischer, G. Z., Soneji, Y., Beinke, S., Salmeron, A., Allen, H., Hay, R. T., Ben-Neriah, Y. and Ley, S. C. (2003) betaTrCP- mediated proteolysis of NF-kappaBl p105 requires phosphorylation of p105 serines 927 and 932. Molecular and cellular biology. 23, 402-413 Lakshmanan, U. and Porter, A. G. (2007) Caspase-4 interacts with TNF receptor-associated factor 6 and mediates lipopolysaccharide-induced NF- kappaB-dependent production of IL-8 and CC chemokine ligand 4 (macrophage-inflammatory protein-1 ). J Immunol. 179, 8480-8490 Pronin, A. N., Morris, A. J., Surguchov, A. and Benovic, J. L. (2000) Synucleins are a novel class of substrates for G protein-coupled receptor kinases. The Journal of biological chemistry. 275, 26515-26522 Carman, C. V., Barak, L. S., Chen, C., Liu-Chen, L. Y., Onorato, J. J., Kennedy, S. P., Caron, M. G. and Benovic, J. L. (2000) Mutational analysis of Gbetagamma and phospholipid interaction with G protein-coupled receptor kinase 2. The Journal of biological chemistry. 275, 10443-10452 Ghoda, L., Lin, X. and Greene, W. C. (1997) The 90-kDa ribosomal S6 kinase (pp90rsk) phosphorylates the N-terminal regulatory domain of lkappaBalpha and stimulates its degradation in vitro. The Journal of biological chemistry. 272, 21281-21288 Taylor, J. A., Bren, G. D, Pennington, K. N., Trushin, S. A., Asin, S. and Paya, C. V. (1999) Serine 32 and serine 36 of lkappaBalpha are directly phosphorylated by protein kinase CKII in vitro. Journal of molecular biology. 290, 839-850 Weller, M., Virrnaux, N. and Mandel, P. (1975) Light-stimulated phosphorylation of rhodopsin in the retina: the presence of a protein kinase that is specific for photobleached rhodopsin. Proceedings of the National Academy of Sciences of the United States of America. 72, 381-385 Benovic, J. L., Strasser, R. H., Caron, M. G. and Lefkowitz, R. J. (1986) Beta- adrenergic receptor kinase: identification of a novel protein kinase that 136 36 37 38 39 4O 41 42 phosphorylates the agonist-occupied form of the receptor. Proceedings of the National Academy of Sciences of the United States of America. 83, 2797-2801 Jimenez-Sainz, M. C., Murga, C., Kavelaars, A., Jurado-Pueyo, M., Krakstad, B. F., Heijnen, C. J., Mayor, F., Jr. and Aragay, A. M. (2006) G protein- coupled receptor kinase 2 negatively regulates chemokine signaling at a level downstream from G protein subunits. Molecular biology of the cell. 17, 25-31 Naga Prasad, S. V., Barak, L. S., Rapacciuolo, A., Caron, M. G. and Rockman, H. A. (2001) Agonist-dependent recruitment of phosphoinositide 3-kinase to the membrane by beta-adrenergic receptor kinase 1. A role in receptor sequestration. The Journal of biological chemistry. 276, 18953-18959 Liu, S., Premont, R. T., Kontos, C. D., Zhu, S. and Rockey, D. C. (2005) A crucial role for GRK2 in regulation of endothelial cell nitric oxide synthase function in portal hypertension. Nature medicine. 11, 952-958 Premont, R. T., Claing, A., Vitale, N., Freeman, J. L., Pitcher, J. A., Patton, W. A., Moss, J., Vaughan, M. and Leflcowitz, R. J. (1998) betaZ-Adrenergic receptor regulation by GIT], a G protein-coupled receptor kinase-associated ADP ribosylation factor GTPase-activating protein. Proceedings of the National Academy of Sciences of the United States of America. 95, 14082- 14087 Martini, J. S., Raake, P., Vinge, L. E., DeGeorge, B., Jr., Chuprun, J. K., Harris, D. M., Gao, E., Eckhart, A. D, Pitcher, J. A. and Koch, W. J. (2008) Uncovering G protein-coupled receptor kinase-5 as a histone deacetylase kinase in the nucleus of cardiomyocytes. Proceedings of the National Academy of Sciences of the United States of America. 105, 12457-12462 Sorriento, D., Ciccarelli, M., Santulli, G., Campanile, A., Altobelli, G. G., Cimini, V., Galasso, G., Astone, D., Piscione, F., Pastore, L., Trimarco, B. and Iaccarino, G. (2008) The G-protein-coupled receptor kinase 5 inhibits NFkappaB transcriptional activity by inducing nuclear accumulation of IkappaB alpha. Proceedings of the National Academy of Sciences of the United States of America. 105, 17818-17823 Kawamata, Y., Irnamura, T., Babendure, J. L., Lu, J. C., Yoshizaki, T. and Olefsky, J. M. (2007) Tumor necrosis factor receptor-1 can function through a G alpha q/ll-beta-arrestin-l signaling complex. The Journal of biological chemistry. 282, 28549-28556 137 43 44 45 46 47 48 Khoa, N. D., Postow, M., Danielsson, J. and Cronstein, B. N. (2006) Tumor necrosis factor-alpha prevents desensitization of Galphas-coupled receptors by regulating GRK2 association with the plasma membrane. Molecular pharmacology. 69, 131 1-1319 Sachdev, S., Hoffmann, A. and Hannink, M. (1998) Nuclear localization of IkappaB alpha is mediated by the second ankyrin repeat: the IkappaB alpha ankyrin repeats define a novel class of cis-acting nuclear import sequences. Molecular and cellular biology. 18, 2524-2534 Legler, D. F ., Micheau, 0., Doucey, M. A., Tschopp, J. and Bron, C. (2003) Recruitment of TNF receptor 1 to lipid rafts is essential for TNFalpha- mediated NF-kappaB activation. Immunity. 18, 65 5-664 Zandi, B, Chen, Y. and Karin, M. (1998) Direct phosphorylation of IkappaB by IKKalpha and H(Kbeta: discrimination between free and NF-kappaB-bound substrate. Science (New York, NY. 281, 1360-1363 Andreakos, E., Smith, C., Kiriakidis, 8., Monaco, C., de Martin, R., Brennan, F. M., Paleolog, E., Feldmann, M. and Foxwell, B. M. (2003) Heterogeneous requirement of IkappaB kinase 2 for inflammatory cytokine and matrix metalloproteinase production in rheumatoid arthritis: implications for therapy. Arthritis and rheumatism. 48, 1901-1912 DiDonato, J. A., Hayakawa, M., Rothwarf, D. M., Zandi, E. and Karin, M. (1997) A cytokine-responsive IkappaB kinase that activates the transcription factor NF-kappaB. Nature. 388, 548-554 138 CHAPTER 4 G-protein coupled receptor kinase 5 (GRK5) mediates Toll-like receptor-4-induced NF-KB pathway in macrophages and is necessary for the production of inflammatory cytokines and chemokines in viva The authors include: Sonika Patial, Shipra Shahi, Yogesh Saini, Daniel M. Appledom, John J. Lapres, Andrea Amalfitano, Narayanan Parameswaran 139 ABSTRACT G-protein coupled receptor kinase-5 (GRK5), a serine-threonine kinase is one of the seven GRK family members, the primary function of which is to regulate the desensitization of G-protein coupled receptors. Recent studies have shown that GRK5 also regulates NFKB pathway stimulated by non-GPCRs. This study was undertaken to determine the potential role of GRK5 in inflammatory signaling in primary . -/- . . . . . . macrophages and in vivo usrng GRK5 mice. Consrstent With our prevrous findings in macrophage cell lines, we demonstrate here that TLR4-induced IKBa-NFKB pathway is inhibited in primary macrophages from GRK5-L mice compared to cells from / . . . . . . GRK5+ + mice. Our results also indicate that this role of GRK5 1S specrfic for IKBa- NFKB pathway because activation of other signaling pathways, including ERK, JNK and p38 are similar between the two genotypes. Consistent with the effects on the NFKB pathway, LPS-induced cytokine/chemokine production was broadly inhibited in -/- . . , . . the cells from GRK5 mice. To examrne the m vzvo relevance of these findings, we injected GRKSJ- and GRK5“+ mice with LPS and measured plasma cytokine levels at various times after injection. Confirming the in vitro data, LPS-induced cytokines/chemokines were significantly inhibited in vivo, in the GRK5"' mice. Associated with these effects LPS-induced liver injury is also decreased in the GRK5"' mice. Taken together, our findings demonstrate that GRK5 acts as a positive regulator of LPS-induced inflammatory signaling and further suggest that these findings could have potential implications for drug development in inflammatory diseases. 140 INTRODUCTION G-protein coupled receptor kinases (GRKs) are serine-threonine protein kinases that regulate the phosphorylation and desensitization of G-protein coupled receptors [1]. GRK family (seven members identified, GRKl-7) is subdivided into three main groups on the basis of sequence homology viz. rhodopsin kinase (GRKI and GRK7), B-adrenergic receptor kinase (GRK2 and GRK3), and the GRK4 kinase (GRK4, GRK5 and GRK6) subfamilies. Although these seven members share certain characteristic features, they are distinct enzymes with specific properties. GRK5 is the best characterized member of the GRK4 subfamily of GRKs which is expressed ubiquitously in all mammalian tissues. It is a membrane associated protein which has been shown to be selectively required for muscarinic receptor desensitization [2]. Recent studies have also shown a nuclear role for the GRK4 family of kinases, owing to the presence of a “Nuclear localization signal” in their sequence[1, 3]. Apart from their role in receptor desensitization, recent studies showed that GRKs also perform other cellular functions by phosphorylating non-receptor substrates such as tubulin, and synucleins [4, 5]. GRKs also interact with a variety of other cellular proteins such as caveolin, calmodulin and actin [6-8]. Along the same lines, recently, GRK5 was also found to interact with members of the IKB (Inhibitor of KB) family. In this regard, GRK5 was shown to interact with and phosphorylate NFKBl p105 as well as IKBa [9, 10]. The fimctional outcomes of these studies were primarily studied using macrophage cell line. Role of GRK5 in regulating NFKB pathway in primary macrophages and the functional relevance of this regulation in vivo, however, is presently not known. 141 NF-KB family of transcription factors (NFKBlpSO, NFKB2p52, RelA(p65), RelB, c-Rel) play an essential role in regulating both innate and adaptive immunity. These proteins are held in the cytoplasm under unstimulated conditions in association with an inhibitory protein family called IKB (0t, B, 8, p100, p105, Bel-3) {reviewed in [11]}. In the canonical NFKB pathway, stimulation with inducers such as LPS or TNFa activates IKB kinase complex, which, phosphorylates IKBa as well as other IKBs including p105. Phosphorylated IKBa undergoes proteasomal degradation thus allowing NFKB factors to translocate to the nucleus and bind onto their cognate DNA binding sites thus initiating the transcription of a wide array of genes including those of cytokines and chemokines. NFKB, although essential for regulating both innate and adaptive immune responses, its constitutive activation is often associated with several inflammatory diseases such as sepsis, rheumatoid arthritis, inflammatory bowel disease, multiple sclerosis, asthma and many more. Because of this role in inflammatory diseases, IKKB has been targeted for drug development, but has faced some pitfalls because inhibition of IKKB not only prevents the deleterious effects of NFKB pathway, it also blocks the protective effects of the NFKB pathway, thereby tipping the balance between inflammatory and antiinflammatory effects of the NFKB pathway towards the harmful outcomes. Thus further understanding of the regulators of the NFKB pathway is an important area being actively pursued by many research laboratories. Based on our previous studies, we hypothesized that deficiency of GRK5 in mice would result in inhibition of NFKB pathway in response to inflammatory stimuli and that would be associated with inhibition of several cytokines and chemokines both 142 from macrophages and in vivo in mice. We demonstrate here that GRK5 is a critical mediator of TLR4-induced NFKB pathway in primary macrophages and in vivo. Importantly, our results also demonstrate a crucial role for GRK5 in inflammation induced by TLR4 activation in vivo in mice. 143 MATERIALS AND METHODS Materials Protease inhibitor cocktail tablets were from Roche Diagnostics (Indianapolis, IN), Phospho ERK, Phospho p38, Phospho JNK, JNK, NFKBI Phospho P105, phospho-IKBa antibodies were from Cell Signaling Technology (Boston, MA). ERK, NF KBl P105 were from Santa Cruz Biotechnology. Tubulin antibody was from sigma. Monoclonal GRK5 antibody was from Upstate Biotechnology. E. coli LPS (0111:B4) from Sigma was used for mice injections. Ultra pure LPS from Invivogen was used for in vitro peritoneal macrophage stimulation. Animals Heterozygous GRK5 mice (backcrossed to C57BL6 background for at least 5 generations) were purchased from Jackson labs. Heterozygous mice were bred to obtain wild type and homozygous GRK5 knockout mice. The litter mate wild types and knockouts were fiirther bred and the F1 and F2 wild type and knockout mice were used for the experiments. Animals were housed four to five mice per cage at 22—24°C in rooms with 50% humidity and a 12-h light—dark cycle. All animals were given mouse chow and water ad libitum. All animal procedures were approved by the Michigan State University Institutional Animal Care and Use Committee and conformed to NIH guidelines. Tail tips were used for isolating genomic DNA and genotyping performed by PCR. All experiments were performed on female mice, 6-8 weeks of age. Peritoneal Macrophage isolation 144 To isolate peritoneal macrophages, mice were injected by intra-peritoneal injection with lml of 4% thioglycollate. Peritoneal macrophages were collected by performing a peritoneal cavity lavage after 4 days of thioglycollate injection in Dulbecco’s phosphate buffered saline (DPBS). Cells were washed atleast three times and then counted and plated on cell culture plates in RPMI 1640 media supplemented with 10% fetal bovine serum (Invitrogen) and penicillin (100 units/ml) and streptomycin (100 pg/ml) at 37° C in 5% C02. After around 18 hours of plating, cells were serum starved for ~ 3-4 hours and stimulated with LPS (1 pg/ml) for the indicated time points. Cytokine analysis A mouse 23-plex multiplex based assay was used to determine the cytokine/chemokine concentrations according to manufacturer’s instructions via Luminex 100 technology as described previously [12]. Plasma from LPS injected mice collected at different time intervals and supematants from peritoneal macrophages stimulated with LPS for different time points were used to assess the cytokine/chemokine levels. Western blot analysis Cells were lysed in lysis buffer (20mM Tris-HCl (pH 7.4), lmM EDTA, 150mM NaCl) containing 1% Triton X-100 with protease inhibitors. Lysed cells were then centrifuged at a maximum speed (13,000 X g) for 10 min at 4°C and protein concentration of the supematants determined by Bradford assay. Western blotting was performed as described previously [9]. Briefly, equal amounts of protein were run on polyacrylamide gels and transferred onto nitrocellulose membranes. The membranes 145 were then blocked in Licor blocking buffer (Licor Biosciences) or 5% w/v skimmed milk for 1 hour after which the membranes were incubated in primary antibodies overnight. Secondary antibodies used were either fluorescently tagged or HRP- conjugated. Blots were developed either on Licor odyssey or using chemiluminescence. Statistical analysis All values are represented as mean 5: SEM. Data were analyzed and statistics performed using GRAPHPAD PRISM software (San Diego, California). The student’s t-test was used to compare mean values between two experimental groups and Analysis of Variance (ANOVA) with Bonferroni post test was used to compare more than two groups. P value of less than 0.05 was considered significant. 146 RESULTS Reduced NFKB activation in GRK5 knockout macrophages Stimulation of cells with LPS and other cytokines particularly TNFa and IL-lB is known to activate IKBOt-NFKB pathway, which plays an essential role in controlling both innate and adaptive immune responses by regulating the transcription of a large number of genes including cytokines and chemokines. As described before, under resting cellular conditions, NFKB is retained in the cytosol by the inhibitory protein IKBa. However, stimulation with LPS triggers the phosphorylation of IKBoi by IKB kinase-B (H{KB). Phosphorylated IKBa then undergoes proteasomal degradation releasing NFKB subunits p50 and p65, which now translocate into the nucleus, bind to its cognate DNA sequence and evoke gene transcription. Previous studies from our laboratory have shown that IKBa interacts with GRK5 and that GRK5 phosphorylates IKBa at least at one of the sites (serine32) phosphorylated by IKKB. In Raw264.7 macrophages cell line, we demonstrated that depletion of GRK5 levels significantly blocks TNFa-induced IKBOL phosphorylation [9]. Based on these results, we proposed that GRK5 interaction with and phosphorylation of IKBOL mediates TNFa-induced NFKB activation in this cell line model. Sorriento et al, at the same time published that GRK5 interaction with IKBOL in endothelial cells, negatively regulates LPS-induced IKBa phosphorylation. These studies found that this effect of GRK5 was independent of its kinase activity [13]. In previous studies, LPS-induced p105 phosphorylation was shown to be negatively regulated by GRK5 in Raw264.7 macrophages cell line [10]. Because of these various observations, we set out to examine the role of GRK5 in NFKB signaling in physiologically relevant cells. For that, we obtained thioglycollate- 147 elicited primary peritoneal macrophages from GRK5“+ and GRK5-L mice and initially tested the effect of LPS on NFKB signaling. We first determined phosphorylation of IKBa in GRK5+/+ and GRK5-L macrophages in response to LPS. IKBa was not phosphorylated either in GRKSJr/+ and GRKSJ- macrophages under basal conditions. Treatment with LPS however caused a marked increase in phosphorylation of IKBa, which peaked at 60 min post stimulation in GRK5“+ cells. Consistent with our previous findings, in GRK5-L cells, IKBa phosphorylation was significantly inhibited (WT:100i0.000%; KO:49.7423:10.907%) (p < 0.001) as compared to GRKSH+ control cells (Fig. 4.1A). To test the significance of this in terms of NFKB activation, we examined the nuclear translocation of NFKB subunit p65 in response to LPS in GRK5+/+ and GRK5-L macrophages. Confirming our finding on IKBO. phosphorylation, nuclear translocation of p65 was significantly inhibited in the knockout cells compared to the wild type macrophages (Fig. 4.1B). Furthermore, electrophoretic mobility shift assays demonstrate greatly attenuated NFKB binding to its consensus sequence in GRK5-L cells compared to the GRKSIL/+ macrophages (Fig. 4.1C). Taken together, these results suggest that GRK5 positively regulates LPS induced NF KB signaling. Interestingly, LPS-induced phosphorylation of NFKB p105 (another IKB protein) was ~20% higher at all time points in the GRK5 knockout macrophages compared to the wild type cells, but was not statistically significant (Fig. 4.1D). 148 Figure 4.1 GRK5 null peritoneal macrophages show reduced NFKB activation A. Thioglycollate elicited peritoneal macrophages from both GRK5+/+ and GRK5-L mice were stimulated with LPS (lpg/ml) for various time points as indicated. Cell lysates were extracted and separated by SDS/PAGE, transferred to nitrocellulose membrane and immunoblotted with primary antibodies against phospho-IKBa and tubulin. Secondary antibodies were either HRP labeled or fluorescent tagged and the blots were developed by chemiluminescence or using LI-COR Biosciences Odyssey system respectively. Representative blots for PIKBa and tubulin and quantification is shown in A. ***p<0.001, N=4. B. Peritoneal macrophages were stimulated with LPS as above and nuclear extracts were prepared and immunoblotted for NF KB p65 (Rel A) as shown. Actin is shown as a loading control. A representative blot from three such experiments is shown. C. Nuclear extracts were also incubated with IRDye 700-labelled NFKB oligonucleotide probes and an EMSA was performed as described in the materials and methods section. Gels were then scanned on a LI-COR Biosciences Odyssey system to detect binding. A representative gel fi'om three such experiments is shown. D. Lysates fi'om peritoneal macrophages stimulated with LPS (lpg/ml) were also immunoblotted using primary antibody against NF KBl p105 (another IKB protein). Representative blot and quantification is shown. Tubulin was used as a loading control. 149 IKBa phosphorylation GRK5 +’* GRK5 "' o 15 30 60 180 o 15 30 so 180 (Time, min) a- plea ~ W Tubulln 1001 80- 0 (D C 8. g 60- 713 .§ 3; 40- E $1 20- Time (min) 150 Figure 4.1. Continued.... GRK5 +’+ GRK5 "' o 30 so 120 o 30 60 120 (Time, min) "his m P65 . .. .. . Actin GRK5 +’+ GRK5 "' o 30 60 120 0 30 so 120 Time, min) 151 Figure 4.1. Continued.... GRK5 *”’ GRK5 "' 0 30 60 120 0 30 so 120 (Time, min) h ‘Oll— M in win-w pNF-KB‘I p105 100. -I-GRK5 +/+ *GRKs-t NFKB1 p105 phosphorylation % maxrmal response 90 120 Time (min) 152 GRK5 does not affect TLR4-induced MAPK activation LPS stimulation of TLR4 also leads to the activation of MAP kinases in addition to NFKB. Because MAPKs (ERK, JN K and p3 8) are also important regulators of inflammatory response and to further determine whether our observed findings on GRK5 regulation of NFKB was specific for that pathway, we sought to examine the LPS-induced activation status of the three important MAPKs in primary macrophages -/- . . . from GRK5 mice and GRK5”+ control mice. Peritoneal macrophages were stimulated with LPS (lpg/ml) for various time points and immunoblotting performed to determine the phosphorylation of MAPKs at different time points. Irnmunoblot analysis however revealed no difference in the kinetics or magnitude of phosphorylation of ERK, JNK and p38 between wild type and GRK5 deficient macrophages suggesting that the GRK5 signaling in response to LPS is selective for IKB-NFKB pathway (Fig. 4.2A, 4.28, 4.2C). Impaired LPS-induced cytokine and chemokine production in GRKS'” peritoneal macrophages To further understand the physiological relevance of our findings, we examined the role of GRK5 in LPS-induced inflammatory cytokine/chemokine production. We hypothesized that because NFKB regulates a number of inflammatory genes, deficiency of GRK5 would broadly inhibit several cytokines/chemokines induced by LPS. For this, we treated primary macrophages from GRK5“+ and GRK 5 mice wrth LPS for various time pornts and collected the cell culture supematants. Cytokine/chemokine levels in the supematants were measured using 23-plex Biorad’s 153 Figure 4.2 GRK5 has no effect on TLR4-induced MAPK activation +/+ Thioglycollate elicited peritoneal macrophages from both GRK5 and GRK5 - mice were stimulated with LPS (lpg/ml) for various time points as indicated. Cell lysates were extracted and separated by SDS/PAGE, transferred to nitrocellulose membrane and immunoblotted with primary antibodies against phospho-ERK, ERK, phospho-JNK, JNK, phospho-p38 and tubulin. Secondary antibodies were fluorescently labeled and the blots were developed using LI-COR Biosciences Odyssey system. (A) P-ERK, ERK and its quantitation is shown. (B) shows a representative blot and quantitation for p-JNK, JNK. (C) show a representative blot and quantitation for p38 and tubulin. N=4. 154 GRK5 *’+ GRK5 4' 0 15 30 60 180 0 15 30 60 180 (Time, min) z: j t “can... pERK +GRK5 H" -A-GRK5 "' 100- .5 31’ 5 § 80- E" 01 £5: 60 CD fig 40- °- «’11 x E E. .\° 2° 0 3'0 6'0 90120150150 Time (min) 155 Figure 4.2. Continued. . .. B GRK5 *’+ GRK5 "' 0 15 30 50 180 0 15 30 so 180 (Time,min) “T " pJNK ~‘flflafiemflmmA-fi “--- ~--“~ JNK 100. '3‘--- I TTTT I ‘i 30.. I; I 60 404 JNK phosphorylation % maximal response o l I I I I T I 0 30 60 90120150180 Time (min) 156 Figure 4.2. Continued.... GRK5 +’+ GRK5 4' 0 31°- 6°. 12°, 0, -w 39 6.0. 120 (Time. min) é'“* m. wail.“ .: ____ .4- _‘ -c. pP38 ~.--—-_.-‘~~TUbUIin +/+ _ +GRK5 -¢-GRK5 /_ p38 phosphorylation % maximal response 0 U I I r 0 30' 60 90 120 Time (min) 157 bioplex assay. As predicted, our results indicate that LPS-induced production of several inflammatory cytokines/chemokines is inhibited in the GRK5 knockout mice macrophages. In particular, after 12 hours. of LPS treatment the following cytokines/chemokines were markedly inhibited: IL-Z (Fig. 4.33, WT: 3.975i0.727pg; KO: 2.285i0.189pg), IL-3 (Fig. 4.3b, WT: 1.680i0.370pg; KO: 0.934i0.136pg), IL-4 (Fig. 4.3c, WT: 5.654i1.233pg; KO: 3.226:I:O.4l9pg), IL-S (Fig. 4.3d, WT: 6.216251.120pg; KO: 3.559zt0.344pg), IL-12p70 (Fig. 4.3c, WT: l9.521d:3.439pg; KO: 11.656zt1.014pg), IL-12p40 (Fig. 4.3f, WT: 195.308zt30.828pg; KO: 105.707i8.911pg), IL-l7 (Fig. 4.3g, WT: 8.6302t2.15213g; KO: 3.129iO.425pg), MCPl (Fig. 4.3b, WT: 691.623i115.893pg; KO: 447.905:l:44.838pg), IFNy (Fig. 4.3i, WT: 12.667:l:2.670pg; KO: 6.710i0.927pg), KC (Fig. 4.3j, WT: 211.4963c40.370pg; KO: 96.345i6.589pg), GM-CSF (Fig. 4.3k, WT: 11.271:I:2.138pg; KO: 6.378i0.570pg), Eotaxin (Fig. 4.31, WT: 91.907i15.865pg; KO: 57.302:Iz5.426pg). An important point to note is that some cytokines/chemokines were similar between the wild type and KO suggesting that GRK5 does not regulate all cytokine/chemokine production en masse. These included TNFa, IL-lB, lL-6, IL-9, IL-13, MIPlB, IL-la, IL-10, GCSF, MIPla, RANTES (data not shown). Impaired LPS signaling in viva in GRK5 knockout mice The secretion of cytokines and chemokines into the plasma is an essential step which orchestrates the anti-infection process. Cytokines and chemokines contribute by enhancing the microbicidal activities of phagocytosing cells such as monocytes and macrophages as well as by recruiting leukocytes to the site of infection. Although contributing towards the anti-infectious process, their excessive production can cause 158 Figure 4.3 LPS induction of cytokines and chemokines in peritoneal macrophages from WT (GRK5+/+) and GRK5 null (GRK5-F) mice Thioglycollate elicited peritoneal macrophages were stimulated with lpg/ml LPS in cell culture plates. Cell culture supematants were then collected at various time intervals and secretary levels of cytokines / chemokines were determined using Biorad’s 23 plex Luminex based assay. Cells were lysed and protein concentration was determined by Bradford assay. Levels of cytokines / chemokines were normalized to total protein concentration and shown in picograrns. Cytokines and chemokines shown include IL-2 (a), IL-3 (b), IL-4, (c), IL-5 (d), IL-12p70 (e), IL-12p40 (f), IL-l7 (g), MCPl (h), IFNY (i), KC (j), GM-CSF (k) and Eotaxin (I). Data was analyzed by Two way ANOVA followed by a Bonferroni posttest for each group. N=6. *p<0.05; **p<0.01; ***p<0.001. 159 U' lL- 3 secretion (pg) lL- 2 secretion (pg) b.’ ‘1’ '1‘ A J Time (Hours) *GRK5*’* . -A-GRK5"' Time (Hours) 160 Figure 4.3. Continued.... C 3‘ *GRK5*’* -A-GRK5"' IL- 4 secretion (pg) Time (Hours) 8. *GRK5*’* I vex-GRK5" IL- 5 secretion (pg) 0 5 1'2 1'3 24 Time (Hours) 161 Figure 4.3. Continued.... +1-1- 25- ‘I'GRKS -A-GRK5"' * A e my 5 mu 5 2 1 1 63 53812.3 N. 1.. 1'5 12 Time (Hours) ‘— 24 H. +4 55 KK RR an {a nw n.. no mu n.. mu 4 0 6 2 8 4 2 2 1 1 33 55.58 see 2 1.. 1'8 12 Time (Hours) 162 Figure 4.3. Continued. . .. 9 14- *GRK5*’* -A-GRK54' IA—‘L 9 '9 IL- 17 secretion (pg) h Time (Hours) 1000' -I- G RK5+I+ 900- 'A- G RK5"' 8001 700- 600- 500- 400- 300- 200- 100- MCP1 secretion (pg) o is 1'2 1'8 24 Time (Hours) 163 Figure 4.3. Continued. . .. —. KC secretion (pg) lFNy secretion (pg) 18- 15‘ 12- 3001 250- 200' 1 501 1 00- 50- 'I'GRK5""' ’A'GRKSJ' 1'3 2'4 Time (Hours) *GRK5*’* 'A'GRKS'” é 1'2 1'3 2'4 Time (Hours) 164 Figure 4.3. Continued. . .. 4 2 *GRK5*’* “A'GRKS ' ‘E__..--........ 1'8 1'2 Time (Hours) 16- k 35 559.03 “50.5.6 14+ .1. ‘I'GRKS 'A'GRKS 1251 3.3 5:203 Exflom Time (Hours) 165 severe side effects. Detectable levels of these inflammatory mediators i.e. cytokines and chemokines in the blood stream are indicative of their exacerbated production. Plasma is one of the major sources which can be utilized to measure the levels of these inflammatory mediators. Moreover, evaluation of plasma cytokines over a time course can provide a better understanding of the nature and severity of the disease process. To investigate the role of GRK5 in TLR4 signaling in vivo, we sought to determine the LPS induced production of cytokines and chemokines in mice plasma. For this purpose, GRKSH+ and GRK5'/' mice were intra-peritoneally injected with LPS (30pg/gm body weight) and blood collected at 1, 3 and 12 hours after injection. Using Biorad’s 23-plex cytokine assay, we determined the plasma levels of various cytokines and chemokines after LPS injection. We rationalized that, because GRK5 mediates a broad spectrum of cytokine/chemokines in the primary macrophages in vitro, LPS injection in vivo in mice would result in diminished cytokine responses in the GRK5"' mice. Consistent with this prediction, LPS-induced cytokine/chemokine levels were broadly inhibited in the GRK5"' mice compared to GRK5”. Again, as is the case with the primary macrophages, not all cytokine/chemokines were affected in this manner. Only a subset of cytokines/chemokines were affected by GRK5 deficiency suggesting selective regulation of these cytokines/chemokines by GRK5. Cytokines that were inhibited in the GRK5"’ mice included TNFa (Fig. 4.43, WT: 218576zt35294pg/ml; KO: 119970i2321lpg/ml at 1 h time point), IL-lB (Fig. 4.4b, WT: 2237.9271648.049pg/ml; KO: 1052.435i108.982pg/ml at 12 h time point ), IL-2 (Fig. 4.4c, WT: 54.615i2.745pg/ml; KO: 36.817i2.053pg/m1 at 12 h time point ), IL-3 (Fig. 4.4d, WT: 37.070i6.248pg/ml; KO: 22.973i2.274pg/ml at 12 h time point), IL-4 (Fig. 166 Figure 4.4 Profile of plasma cytokines after LPS challenge in WT (GRK5+/+) and GRK5 null (GRK5-F) mice Plasma cytokines TNFa (a), IL-lB (b), IL-2 (c), IL-3 (d), IL-4 (e), IL-5 (1’), IL-lO (g), IL-12 (p40) (h), IL-12 (p70) (i), IL-13 (j), IL-17 (k), MCPl (I), Rantes (m), Eotaxin (n) were determined in WT and GRK5'/' mice after an intra-peritoneal injection of 30pg/g body weight LPS at specified times viz. 1 hr, 3 hrs and 12 hrs (N=6 per group). Data was analyzed by Two way ANOVA followed by a Bonferroni posttest for each animal group. *p<0.05; **p<0.01; ***p<0.001. 167 ‘P 0) OJ C 0| O O O O n n 25004 N O O O n 1500- 1000- 5004 Plasma IL-1B(pglml) 0' Plasma TNFMPQImI) 9’ 4.0 J 0 are» 3’ 20- 101 Plasma IL- 2 (pg/ml) h D § i i '- GRK5 ”t -A'GRK5 "- *** Noinjctrl 1'hr 3hrs 12hrs Time (Hours) -l- GRK5 *’* -A- GRK5 "' No inj ctrl 1i1rs 3hrs 12'hrs Time (Hours) ‘- GRK5 *’* -A- GRK5 "' *** “‘~23 No in] ctrl 1i1rs sins 12'hrs Time (Hours) 168 Q. «5 0| 0 O l 1 Plasma IL- 3 (pg/ml) o: C Figure 4.4. Continued. . .. fkoRK5+ CD 12.5- 10.0- 7.5' 5.0' 25* Plasma IL- 4 (pg/ml) No in] ctrl 1hrs sins 12'hrs Time (Hours) '- GRK5 *’* -A- GRK5 "' *** ...“i§ 0.0 600- 500- v 400- 300- 200‘ 100- ngmI) '4' Plasma lL- 5 No in] ctrl 1i1rs 3hrs 12'hrs Time (Hours) *** -I- GRK5 t“ -A- GRK5 "- .---i§ No inj ctrl 1hrs 31'1rs 12'hrs Time (Hours) 169 (Q 3' Figure 4.4. Continued.... 20000- * '- GRK5 ”t 10000- Plasma IL- 10 (pg/ml) No inj ctrl 1i|rs 3I'1rs 12'hrs Time (Hours) -l- GRK5 *** -A- GRK5 "' *** N 03 O O O O O O <.= <.= 1ooool Plasma lL- 12 (p40) (pg/ml) No inj ctrl 1 hrs 3 hrs 12'hrs Time (Hours) 400- -I-GRK5 *’* *** 300.. 'A' GRK5 4' A N C O O O n n L Plasma IL- 12 (p70) (pg/ml) No inj ctrl 1 hrs 3 hrs 12'hrs Time (Hours) 170 Figure 4.4. Continued. . .. 3000- 2000‘ 1000- Plasma IL- 13 (pg/ml) h.- -I- GRK5 *’+ *** -A- GRK5 "' x 6000- 50004 40004 3000- 2000- 1000‘ Plasma IL- 17 (pg/ml) No i'njctrl 1hrs 3hrs 12'hrs Time (Hours) ** -I- GRK5 *’* -A- GRK5 "' No inj ctrl 1 hrs 3 hrs 12 hrs Time (Hours) 200000- 100000- Plasma MCP1 (pg/ml) ‘- GRK5 *’* *** fikoRK5+ * Noinjctrl1hrs 3hrs 12'hrs Time (Hours) 171 Figure 4.4. Continued.... m ‘5 20000- 31 -I- GRK5 +’* *** 3 -A'GRK5 "' (D E 2 10000- :15 ----A m E m (B a 04 No inj ctrl 1hrs 31'1rs 12'hrs Time (Hours) 8000- *** Plasma Eotaxin (pg/ml) 3 No inj ctrl 1 hrs 3 hrs 12'hrs Time (Hours) 172 4.4e, WT: 8.970i0.571pg/m1; KO: 5.672i0.375pg/ml at 12 h time point), IL-5 (Fig. 4.4f, WT: 417.9932t129.706pg/ml; KO: 98.120i24.171pg/ml at 12 h time point), IL- 10 (Fig. 4.4g, WT: 13648.600i3891.250pg/ml; KO: 5645.438i1033.724pg/ml at 12 h time point), IL-12p4O (Fig. 4.4b, WT: 22694i3038pg/ml; KO: 5101:}:1129pg/ml at 12 h time point), IL-12p70 (Fig. 4.4i, WT: 288$:40pg/ml; KO: 137i13pg/ml at 12 h time point) IL-l3 (Fig. 4.4i, WT: 2220i163pg/ml; KO: 11581201pg/ml at 12 h time point), IL-17 (Fig. 4.4k, WT: 3780i17S6pg/m1; KO: 195i6lpg/ml at 12 h time point), MCP1 (Fig. 4.41, WT: l39016i38025pg/ml; KO: 36490i12175pg/ml at 12 h time point), Rantes (Fig. 4.4m, WT: 13920i1855pg/ml; KO: 7510i651pg/ml at 12 h time point), Eotaxin (Fig. 4.4n, WT: 6548i615pg/ml; KO: 4443i471pg/ml at 12 h time point). Cytokines that were not affected by GRK5 deficiency included GCSF, GM-CSF, lFNy, IL-la, IL—6, IL—9, KC, MIPla, MIPIB (data not shown). Diminished liver injury in GRK5-l. mice Due to the fact that a major group of “pro-inflammatory” cytokines and chemokines were markedly decreased in GRK5'/' mice, we hypothesized that tissue injury seen as a result of LPS-induced endotoxemia would be reduced in GRK5"' mice. To this end, we assessed the extent of liver damage by determining the plasma concentrations of liver injury marker alanine transaminase (ALT) at 12 hours after LPS challenge, both in control GRK5"' and GRK5 deficient i.e. GRKS'I' mice. Our results show that GRK5'/' mice display significantly reduced levels of plasma ALT as compared to GRK5"+ mice (503:4 U/L in GRK5'/' mice verses 84i10 U/L in GRKS'l' mice) demonstrating that GRK5 deficiency results in reduced LPS dependent 173 Figure 4.5 GRK5-I. mice show reduced LPS-induced liver injury GRKS'L/+ and GRK5'/' mice were injected with LPS and plasma levels of liver injury marker ALT were measured after 12 hours. Data was analyzed by unpaired t test; N=6. *p<0.05 174 EJGRK5”* I GRK5 "' 100 5 O 5 7 5 2 :5. .22 5< 175 production of inflammatory mediators which translates into a decreased liver injury (Fig. 4.5). Taken together, these results suggest that GRK5 plays an important role in LPS mediated signaling pathway affecting crucial transcription factors responsible for the production of a wide array of pro- as well as anti-inflammatory cytokines, not only in primary macrophages in vitro, but also in vivo. 176 DISCUSSION LPS binding onto TLR4 induces the activation of NFKB as well MAPK signaling pathways. In this study we show that GRK5, a GPCR kinase is a regulator of LPS-induced NFKB signaling pathway. GRK5 null primary macrophages show a significant reduction in LPS-induced phosphorylation of IKBO. as well as nuclear translocation of p65, a subunit of NFKB. Furthermore, the binding of NFKB onto its consensus DNA oligonucleotide was also found to be significantly inhibited in GRKS' /' macrophages. These findings are consistent with our recent report that siRNA mediated knockdown of GRK5 in Raw264.7 macrophage cell line significantly blocks TNFa-induced NFKB signaling. In this study we not only confirm our previous findings, we also provide evidence that GRK5, by mediating NFKB signaling, has biologically relevant role, in the production of inflammatory cytokines/chemokines in both primary macrophages in vitro and in mice in vivo. Our studies further show that the MAPKs including ERK1/2, JNK and p38 are not regulated by GRK5. LPS-induced P105-ERK pathway was found to be negatively regulated by GRK5 in Raw264.7 macrophage cell line using RNAi against GRK5 [10]. Although we observed ~20% enhancement of pPlOS and pERK levels in the GRK5"' macrophages, it did not reach statistical significance. Thus, Raw264.7 macrophages are clearly different from primary macrophages with regard to the role of GRK5 in ERK activation. It is also quite possible that the different results obtained may be related to the mechanism by which GRK5 was depleted. Germline deletion of a gene could have different consequences when compared to RNAi knockdown. In particular other compensatory mechanisms can ceme into play when germline deletion is used. 177 Interestingly, with regard to GRK5 regulation of IKBoc, it appears that both methods lead to similar effects suggesting that GRK5 is an important if not the only regulator of IxBa-NFKB signaling. It is important to point out that other kinases including IKKa, casein kinase and ribosomal S6K have also been shown to regulate IKBOL- NFKB signaling by phosphorylating IKBG. [14-16]. It is clear in the GRK5 knockout that other kinases (especially IKKB) are also phosphorylating IKBG. because phosphorylation in the absence of GRK5 was not completely blocked, but was inhibited ~50%. LPS binding onto TLR4 receptor leads to the activation of both NFKB and MAPK signaling that eventually induces the expression and production of several cytokines and chemokines. We found that the secretion of several inflammatory cytokines and chemokines is regulated by GRK5 in peritoneal macrophages as well as under in vivo conditions. LPS induction of TNFa, IL-IB, IL-2, 3, 4, 5, 10, IL-12p40, IL-12p70, IL-13, IL-17, MCP1, Rantes and eotaxin were found to be inhibited in GRK5'/' mouse plasma. Cytokines are soluble mediators that coordinate inflammation by being secreted from one cell and activate receptors on other cells. TNFa is the first cytokine that appears in the blood of experimental animal models after LPS injection as well as in human volunteers receiving LPS [17]. TNFa causes the induction of a large number of inflammatory mediators by acting through a signaling cascade as well as by autocrine mechanisms [18, 19]. This has been shown by blocking TNFOL which caused a decrease in the levels of other cytokines. For e.g. in baboons infected with E.coli, blocking TNFa significantly reduced the levels of IL-la, IL-6 and IL-8 [20, 21]. 178 Concentration of TNFo. observed in sepsis patients correlates with the severity and outcome of the disease. For eg., concentrations of TNFa as well as IL-IB and IL-2 in plasma were found to be higher in patients of septic shock as compared to patients of sepsis alone [22]. In our studies, LPS injection resulted in the secretion of TNFa in the plasma of both GRKS'L/+ and GRK5-L mice at 1 hour post injection. The levels . . . -/- . observed were, however, srgnlficantly less in GRK5 mice. IL-l family of cytokines, in particular IL-lB is a key pro-inflammatory mediator. IL-lB is released in response to an insult and initiates a host defense response by up-regulating other cytokines, acute phase proteins as well as by acting as a potent pyrogen [23, 24]. Furthermore, it has been shown to be required for the clearing of bacterial infections [25]. IL-IB does so by activating MAPK and NFKB pathways [26]. Excessive activation of IL-lB however, results in multi organ failure which is commonly observed in sepsis [27]. We found sustained plasma levels of IL- IB up to 12 hours of LPS injection in GRK5+/+ mice. However, IL-IB levels reach a peak by 3 hours and thereby start declining in GRK5'/' mice. At 12 hours, the levels observed in GRK5'/' mice plasma were significantly less as compared to those observed in GRK5++ mice. Initial increase 1n lL-lB rs important to fight agalnst infection, however, a sustained activation might lead to endotoxemia. We did not see a significant difference in the levels of IL-IB in the cell culture supematants of peritoneal macrophages however, suggesting that GRK5 might regulate the production 179 .4 I.“ W“? ***-m of IL-lB in cells other than macrophages. It is also possible that the integrative pathophysiology present in vivo may not be reproducible in vitro. IL-12p40 is produced by activated macrophages, neutrophils, microglia and dendritic cells. IL-12 has been shown to be important for the production of IFNy and leads to lethality in LPS induced septic shock model of mice [28]. IL-17 is secreted primarily by T lymphocytes and is involved in recruiting neutrophils and acts together with other cytokines such as TNFa and IL-IB, to further enhance inflammation [29]. Eotaxin and MCP1 act as chemoattractants for eosinphils and leucocytes respectively. IL-13, IL-4 and IL-10 are anti—inflammatory cytokines which act by inhibiting the production of pro-inflammatory cytokines. For instance, IL-l3 has been shown to act by inhibiting the production of IL-IB and TNFa which are induced by LPS. Since some of the cytokines which were found to be reduced in GRK5'/' mice are primarily T-cell cytokines, fiiture studies will look into the role of these cell type specific cytokines in inducing inflammation. Although secretion of pro-inflammatory cytokines serves as an essential prerequisite for initiating an effective innate immune response to fight infection, they are also associated with deleterious effects leading to multi organ failure and ultimately death. Similarly, anti-inflammatory cytokines despite important for controlling an exaggerated inflammatory response, also lead to a suppression of the immune system which is required for a proper functioning of the system. Systemic inflammatory response syndrome (SIRS) is a consequence of an imbalance between pro and anti-inflammatory cytokines. Our studies show that absence of GRK5 reduces the production of several pro as well as anti-inflammatory cytokines. These cytokines 180 are still produced to some extent which is crucial for inducing an effective immune response. However, since exaggerated production of these inflammatory mediators is modulated by GRK5, we speculate that our results would have important consequences in inflammatory diseases. In fact, our observations on reduced ALT levels in the GRK5—l- mice compared to the GRK5+/+ mice suggests that inhibition of GRK5 in sepsis might mitigate organ injury. Future studies will address whether GRK5 would be a viable therapeutic target for inflammatory diseases. 181 REFERENCES 1 Johnson, L. R., Scott, M. G. and Pitcher, J. A. (2004) G protein-coupled receptor kinase 5 contains a DNA-binding nuclear localization sequence. Mol Cell Biol. 24, 10169-10179 2 Gainetdinov, R. R., Bohn, L. M., Walker, J. K., Laporte, S. A., Macrae, A. D., Caron, M. G., Lefl(owitz, R. J. and Premont, R. T. (1999) Muscarinic supersensitivity and impaired receptor desensitization in G protein-coupled receptor kinase 5-deficient mice. Neuron. 24, 1029-1036 3 Martini, J. S., Raake, P., Vinge, L. E., DeGeorge, B. R., Jr., Chuprun, J. K., Harris, D. M., Gao, E., Eckhart, A. D., Pitcher, J. A. and Koch, W. J. (2008) Uncovering G protein-coupled receptor kinase-5 as a histone deacetylase kinase in the nucleus of cardiomyocytes. Proc Natl Acad Sci U S A. 105, 12457-12462 4 Haga, K., Ogawa, H., Haga, T. and Murofushi, H. (1998) GTP-binding- protein-coupled receptor kinase 2 (GRK2) binds and phosphorylates tubulin. Eur J Biochem. 255, 363-368 5 Pronin, A. N., Morris, A. J., Surguchov, A. and Benovic, J. L. (2000) Synucleins are a novel class of substrates for G protein-coupled receptor kinases. J Biol Chem. 275, 26515-26522 6 Carman, C. V., Lisanti, M. P. and Benovic, J. L. (1999) Regulation of G protein-coupled receptor kinases by caveolin. J Biol Chem. 274, 8858-8864 7 Freeman, J. L., De La Cruz, E. M., Pollard, T. D., Lefl ** 0.12- 0.10- ) o b 8" 0.06- BALF #9 I Pl ( o "o i“ 0.021 O'OO'GRK'Z "’" GRK2 “We 12 hour LPS injection 212 m .m m c 4 5 n w. n F I .. and? 2.. 213 Percent survival Figure 5.4. Continued.... C. 100 80- 601 40- 201 0 8 1'2 1'8 2'4 3'0 3'6 4'2 48 Time (Hours) 214 ml Fur—MLJI. " . . . fl inflammatory cytokines VIZ. TNFa (59221247pg in GRKZAmye; 363i60pg 1n GRKZfl/ at 6 h), IL-6 (185i26pg in Grammy"; 80i19pg in GRKZ'M' at 24 h), IL-la Amye (5.8il.lpg in GRK2 ; 3.2i1.1pg in GRK2fl/fl at 24 h), IL-lB (55.2:1:5.3pg in Amye GRK2 ; 27.5:l:4.7pg in GRKzfl/fl at 24 h), IL-9 (24i2pg in GRKZAmye; 111:ng in GRK2fl/fl at 12 h) as well as chemokines viz. MCP1 (155221: 82pg in GRKZAmye; 710i111pg in GRKZfl/fl at 12 h), MIPla (16401178111; in GRKZAmye; 815i136pg in GRKZfl/fl at 24h) and GCSF (117:1:22pg in Grimm”; 47ilOpg in (3111(szfl at 24 h) in the GRK2 deficient mice macrophages compared to control cells. Importantly, cytokines including IL-2, IL-12p40, IFNy and IL-17 did not differ between control and GRK2 deficient mice macrophages, suggesting selective regulation of TLR4- induced cytokine responses by GRK2. To determine the differences between macrophages and neutrophils in terms of the cytokine regulation, we also assessed the levels of cytokines and chemokines in neutrophils (treated in vitro) from these mice. Compared to the results obtained with macrophages we observed that neutrophils from GRK2 deficient mice show an enhanced secretion of only G-CSF and KC compared to control mice (Figure 5.6). Other cytokines such as IL-la, IL-18, IL-6, IL-12 (p70), GMCSF, MIPla, and MIPIB showed a similar trend although these were not statistically significant. Furthermore, some cytokine/chemokines (IL-12p40, RANTES, IL-17) were not different between control and GRK2 deficient mice suggesting specific regulation of some cytokines by GRK2 in the neutrophils. GRK2 deficiency augments LPS-stimulated ERK phosphorylation but not P38 and JNK phosphorylation in primary macrophages 215 Figure 5.5 Peritoneal macrophages from GRK2Amye mice secrete enhanced amounts of pro-inflammatory cytokines and chemokines in response to LPS stimulation Thioglycollate-elicited peritoneal macrophages were stimulated with LPS (lug/ml) in 12-well cell culture plates and cell culture medium was collected at different time intervals. Levels of inflammatory mediators were determined using biorad 23 plex assay. Cells were lysed to determine the protein concentration using Bradford method. Levels of TNFa, IL-6, IL-la, IL-18, IL-9, MCP1, MIPla and GCSF are shown as pg/iig of total cellular protein. *p<0.05; **p<0.01; ***p<0.001. 216 P2.. V“ TNFa secretion in pg lL-1a secretion in pg q .- fol q 800 -A- SEE; Amye 225 -I- G RKZ fl/fl *** 700 1 * 2001 -A- GRK2 Amye l : “ o' .E 1 0" 500 2 : 0‘ g 5 : .o . 125- 400 '1 : “ '0' g : \‘ ',' 8 100‘ 300 - : "E ,o' a : ‘ ' ‘9 75‘ 1001 25- 0 I V I W e I I I I 0 6 12 18 24 0 6 12 18 24 Time (Hours) Time (Hours) 7- 'I'GRKZ “’1" * so. *GRKZ W“ *** 'A' GRK2 Amye l 'A' GRK2 Amye I 6" a ' ' o ' a 50' *1: .1" 5- .5 Z" ,- g 40- .- . 4. a x . . 31 in '0 m. o s- 20" ' 2- A i 1 . 10" 'I' c . . . . 0L. . - . . 0 6 12 18 24 0 6 12 18 24 Time (Hours) 217 Time (Hours) Figure 5.5 Continued... Time (Hours) 451 'I'GRKZ W" 4% ‘A'GRKZ Me I g 35- ' 83 C .E 30. 'E c .o i i s 20- 8 w .- o: 151 ii :£ 10. E; 511 c I T I I 0 6 12 18 24 Time (Hours) 225“ 'l'GRKZ "I" 2000- 'A'GRKZ Amye m 83 l 83 .5 ml ------------- .s C C a e 2 2 ii i? 2 J 11 a- ‘6’ 2 :9 1'8 2'4 218 *GRKZ fl’" *** 'A'GRKZ We .5 gs 1 501 1 251 1 001 Time (Hours) 'I'GRK2 W" *** ’A'GRKZ “We 1 Time (Hours) Figure 5.6 Neutrophils from GRK2Amye mice secrete enhanced amounts of pro-inflammatory cytokines and chemokines in response to LPS stimulation Thioglycollate-elicited neutrophils were stimulated with LPS (lug/ml) in 12- well cell culture plates and cell culture medium was collected at different time intervals. Levels of inflammatory mediators were determined using biorad 23 plex assay. Cells were lysed to determine the protein concentration using Bradford method. Levels of GCSF, KC, IL-la, IL-IB, IL-6, IL-12p70, GM- CSF, MIPla and MIPlB are shown as pg/pg of total cellular protein. **p<0.01. 219 G-CSF secretion (pg) lL-1 B secretion (pg) GM-CSF secretion (pg) 80- *Gszfo'fi'GRKngYG 0 6 1'2 1'8 2'4 Time (Hours) *GRKzfllfl .A.GRK2AMye 201 0 0 6 1'2 1'8 2'4 Time (Hours) 17.5-'.'GRK2fl/fl 'A'GRKZAMye 15.0- 12.5- 10.0- 7.5- 5.0- 2.5- 0.0 0 6 1'2 1'8 2'4 Time (Hours) 220 Figure 5.6 Continued. . .. IL-6 secretion (99) KC secretion (pg) MIP-1 a secretion (pg) 1251 100- l 'I'GRKZflm 'A-GRK2AMYS 75- x 50- '0' 25- 300- 2001 100- 0 6 1'2 1'8 2'4 Time (Hours) 'I'GRKZflm 'A'GRKZAMye 0 6 1'2 1'8 2'4 Time (Hours) 200- 100- O 0 6 1'2 1'8 2'4 Time (Hours) 221 Figure 5.6 Continued. . .. lL-12p70 secretion (pg) NIP-1 (3 secretion (pg) 51 -I-GRK2fl/fl .A.GRK2AMye lL-1 a secretion (pg) (.0 0 6 1'2 1'8 2'4 Time (Hours) 17.51 *GRKZ'V'I 'A'GRKZAMye 15.01 12.51 10.01 7.5- 5.0- 2.51 0.0 0 6 1'2 1'8 2'4 Time (Hours) 1751'GRK2fl/fl 'A’GRKZAMye 1501 1251 1001 751 50- 251 0 6 1'2 1'8 24 Time (Hours) 222 ‘ . ‘. A" sou-(aw FA To begin to understand the mechanisms of how GRK2 negatively regulates LPS-induced cytokine/chemokine response in vivo and in primary macrophages in vitro, we tested the effect of LPS in GRK2 deficient and control cells on various signaling pathways. LPS stimulation of TLR4 signaling leads to the activation of MAPK signaling cascades including ERK, JNK and p38 kinases as well as IKBa- NFKB signaling pathways. These pathways have been shown to be the central regulators of inflammatory responses in endotoxemia. Therefore, we assessed the phosphorylation status of ERK, JNK, p38 and IKBa in the primary macrophages both in control as well as GRK2 deficient macrophages upon exposure to LPS. Peritoneal macrophages from GRK2 deficient and control mice were stimulated with LPS for various time points and immunoblotting performed for pERK1/2, pJNK, pP38 and pIicBa. Our results suggest an interestingly selective role for GRK2 in LPS-induced ERK phosphorylation. Thus, although LPS-stimulated pJNK, pP38 and pIKBa levels were similar between the control and GRK2 deficient macrophages (Fig. 5.7B), pERK levels were markedly enhanced in the GRK2 deficient cells (Figure 5.7A). Phospho- ERK levels were 36i11% and 95i3% at 15 and 30 min respectively in GRK2 deficient cells, whereas in control cells the levels reached only 13i3% and 51i8% at 15 and 30 min respectively. We also examined the levels of pAkt and pGSK3 (known to be regulated by GRK2) after LPS stimulation and did not find any evidence for their regulation in these cells by GRK2 (Figure 5.8). GRK2 negatively regulates NF—KBI-plOS-TPLZ-MEK-ERK pathway in primary macrophages 223 Figure 5.7 GRK2 deficiency causes an enhanced LPS stimulated ERK1/2 activation in peritoneal macrophages A. GRK2Amye and control primary peritoneal macrophages were stimulated with lug/ml of LPS for different time points and the phosphorylation of MAPKs and IKBO. was determined by western blotting. A representative blot for phospho ERK and ERK and quantification for the same is shown in (A). ***p<0.001 compared to control. N=6. B. A representative blot for phospho-p38, actin, phospho-JNK, JNK and phospho-IKBa, tubulin is shown. 224 A. GRKzflm GRKZAmye 0 15 30 60 90 120 180 0 15 30 60 90 120180 (Time, min) :: $3 1': ' ' 2: 352 '1": pERK ~~~~~~~~ 1- "- 1-- -- - - ERK 125 +6sz "l“ . . Amye *** A GRK2 —\ O O \l 0| 0| 0 25 ERK phosphorylation (% maximal response) O 30 60 90 120150180 Time (min) 225 Figure 5.7. Continued.... GRK2 fllfl GRK2 Amye 0 15 30 60 0 15 30 60 (Time,min) I - I: f. 1:1. .- ., .u. ~ .5_ .1 _ ,. 1... , . ,.. ; 1 ‘ ’13.." ‘ "W . ‘12:, ."‘7"."'-:. ?”.'E:, . '2" 5.12;. 11.11:) mains» steals-4.5.5» east-5558.911} flexes-.1982! Infra-1.1118381? 4?? i.» I. .6 i -. ACtI n iii? “1",", “-11 - 111 111° W.._-JNK 111 . _ ..._T‘:'-lP|KBa ’H‘ ' I *" In.‘ 1...:- 1. F1” mun w “,2... 1"" TUbUIIn 226 *4? Figure 5.8 GRK2 deficiency does not affect LPS stimulation of Akt and GSK3|1 activation in peritoneal macrophages GRKZAmye and control primary peritoneal macrophages were stimulated with lug/ml of LPS for different time points and the phosphorylation of Akt and GSK3B was determined by western blotting. A representative blot for phospho-Akt, Akt, phospho-GSK3B and tubulin is shown. 227 GRK2 "I" GRK2 Amve 0 15 30 60 0 15 30 60 (Time, min) pAkt ~~fl~_~'“”Akt GRK2 "m GRK2 MW" 0 15 30 60 0 15 30 60 (Time, min) “pGSK3 it. n— a... :g-mgggm—grubulin 228 To further elucidate the biochemical mechanisms of GRK2 regulation of the ERK pathway, we examined the upstream regulators of ERK phosphorylation in primary macrophages. Previous studies have demonstrated that ERK activation in macrophages in uniquely regulated via LPS-stimulated IKKB-NFKB] p105 pathway. That is, under unstimulated conditions, p105 (an IKB family member bound to NFKB p50 subunits) is also stoichiometrically bound to TPL2 (a MAP3K). When activated, p105 is phosphorylated by IKKB, which then undergoes partial degradation releasing NFKB p50 subunits as well as TPL2. TPL2 then phosphorylates and activates MEK1/2, which then activates ERK1/2. We first confirmed the existence of IKKB mediated ERK signaling pathway in peritoneal macrophages. For this purpose, primary peritoneal macrophages from GRK2 control mice were stimulated with either LPS alone or pretreated with BMS345541 (IKKB inhibitor) before stimulation with LPS and the phosphorylation of ERK was assessed by immumoblotting. As shown in Figure 5.9, we observed that the pharmacological inhibition of IKKB indeed attenuated ERK phosphorylation confirming the existence of IKKB-NFKB] p105 / TPL2-MEK-ERK pathway. To investigate at what level GRK2 regulates this pathway, we examined the phosphorylation of MEK1/2 and p105 after LPS stimulation in control and GRK2 deficient macrophages. Our studies reveal that GRK2 negatively regulates ERK pathway at the level of p105, because LPS-induced MEK1/2 as well as p105 phosphorylation were both enhanced significantly in the GRK2 deficient macrophages compared to control cells (Figure 5.10). Phospho-p105 levels were 471:1 5% and 84i9% at 15 and 30 min respectively after LPS treatment in the GRK2 229 Figure 5.9 The occurrence of IKKB-NFKBlp105/T plZ-MEK-ERK pathway in peritoneal macrophages confirmed by using an IKKB inhibitor Peritoneal macrophages from control mice were stimulated with either LPS alone or pretreated with BMS345541 (SpM), a specific inhibitor against IKKB, 30 minutes prior to stimulation with LPS (lug/ml) for various time points as shown. Irnmunoblotting was performed for phospho-ERK and ERK proteins using specific antibodies. A representative blot is shown. 230 l [ LPS LPS+IKKi 0 30 60 0 30 60 (Time, min) 5:: ....;: pERK 231 deficient cells compared to 21i5% and 5321:10% at 15 and 30 min respectively in control cells. Because LPS-induced IKKB-mediated IKBO. phosphorylation was not affected by GRK2, our results indicate that GRK2 likely regulates at the level of p105. This is further supported by previous studies, which have shown that GRK2 indeed directly interacts with p105, even though these studies did not find any functional. significance of this interaction. Taken together our studies indicate that GRK2 interaction with p105 negatively regulates LPS-induced ERK activation, as well as potentially the p50-mediated NFKB activation. Role of enhanced p105-ERK activation on exaggerated inflammatory response in GRK2 deficient mice Our results so far suggest that deficiency of GRK2 in primary macrophages results in the enhanced activation of p105-ERK pathway, which is associated with an enhanced inflammatory cytokine response observed in macrophages as well as in vivo in mice. To further demonstrate that the effects on the cytokines observed in the GRK2 deficient mice are a result of enhanced p105-ERK activation, we tested the effects of LPS on cytokine/chemokine responses in primary macrophages in presence or absence of an ERK inhibitor. We treated peritoneal macrophages from control and GRK2 deficient mice with LPS in the presence or absence of PD98059 (a MEK inhibitor). Cytokine/chemokine secretion was determined using 23-plex Biorad assay kit. Among the cytokine/chemokines that were enhanced in the GRK2 knockout macrophages, the ERK inhibitor inhibited IL-la, MIPla, GCSF and MCP1 in the GRK2 knockout macrophages but not in the control mice macrophages. In the presence of the ERK inhibitor, the levels cytokines/chemokines returned to the levels 232 .ua‘i’. l Figure 5.10 GRK2 deficiency causes an enhanced LPS stimulated NFKB] p105 activation in peritoneal macrophages GRK2Arnye and control primary peritoneal macrophages were stimulated with 1 jig/ml of LPS for different time points and the activation of NFKB] p105 and MEK were determined by western blotting. A representative blot for phospho- NFKBl p105, ERK, phospho-MEK, tubulin and quantification for the same is shown. ERK and tubulin were used as loading controls. *p<0.05 compared to control. N=7. 233 GRK2 "’1' GRK2 MW" 0 15 30 60 90120180 0 15 30 60 90120 180 (Time,min) . 2.... -,.... ~1- "" "*"‘ ‘ ' NFKB1P105 be... -- in“ Inm- ~ ‘ “M 111-11' = pMEK ~~~...-).L.~.~~~~~muTubulin —.— GRK2 fl/fl .fi. GRK2 Amye NFKB1 p105 phosphorylation (% maxrmal response) 0 0 30 60 90 120150180 Time (min) 234 observe than the lL-la. l GRK2 examine of IKK] also Sig Interest were bl these Iv likely i consislc neither : genotyp ERK pa in reSpQ observed in the control LPS treated cells (except for MIPla which was slightly higher than the control LPS) (Fig. 5.11A). This demonstrates that the enhanced secretion of IL-la, MlPla, GCSF and MCP1 result from an enhanced ERK activation observed in GRK2 deficient macrophages. Because IKK-p105 pathway regulates ERK, we examined if the secretion of these inflammatory factors are also affected by inhibition of IKKB. Except for MCP-l, the other three factors i.e. IL-la, MIPIOL, GCSF were also significantly blocked with the IKKB inhibitor (BMS345541) (data not shown). Interestingly the enhanced secretion of IL-6, and IL-18 in the GRK2 deficient cells were blocked only by the IKKB inhibitor and not by ERK inhibitor, suggesting that these two cytokines are regulated by GRK2 exclusively via the IKKB-p105 pathway likely involving the p50-NFKB activation (Figure 5.11B). Although IL-9 was consistently enhanced in the GRK2 deficient cells compared to the control cells, neither BMS345541 nor PD98059 efficiently blocked IL-9 secretion in cells from both genotypes. Thus IL-9 may not be exclusively regulated by IKKB or ERK. Taken together, our results demonstrate that GRK2 negatively regulates p105- ERK pathway thereby regulating the production of ILl-a, GCSF, MIPIOL and MCP1 in response to LPS. 235 Figure 5.11 Pharmacological inhibition of ERK and IKKB cause a significant inhibition of LPS-induced secretion of cytokines and m e . y mice chemokines in peritoneal macrophages from GRK2A A. Peritoneal macrophages were either left unstimulated, stimulated with LPS or pretreated with PD98059 (lOuM), a MEK inhibitor, 30 minutes prior to stimulation with LPS (lug/ml) and the cell culture supernatant was collected 24 hours later. The secretion of cytokines and chemokines was assessed as described before using biorad 23 plex assay. N=5. *p<0.05; **p<0.01; ***p<0.001. B. Peritoneal macrophages were either left unstimulated, stimulated with LPS or pretreated with BMS345541 (SliM), a specific inhibitor against IKKB, 30 minutes prior to stimulation with LPS (lug/ml) and the cell culture supernatant was collected 24 hours later. The secretion of cytokines and chemokines was assessed as described before using biorad 23 plex assay. N=5. *p<0.05; **p<0.01; ***p<0.001. 236 5.0.5. 207 221 A NS l—I $6 15.01 12.51 10 01 €655 5.2.3 :33 9:9: coweoom 8 Fl: .: .. 1 000- mmmmeemmme 9 8 7 6 w 40.. 3 2 1 9&0 E895 53:3 .23 9:9: :2383 SEE 237 Jill! .~V -. . * * vnwv .. I . V “ch L“. o 99 0V 4‘ 6W. «.6 09 0 9 v9 fl 04¢an gQV QV! 4‘0; 90 90 m. «3? o m 9...“. em. 0.\ $0.\ Q .I. Tl. r§ (V? rl T v a“ [$8 Av ovx 9? e 0.9 9Q $0 » 1.... .33» 3333?? nwumwoeoq nunwnmysz .v m. .5395 3.2.3 .33 9.58 9 .539... 3.2.8 .33 9.3... cozmbumm “50.0 5.553 E05. Figure 5.11. Continued.... 238 *** ** Figure 5.11. Continued.... 301 W00 21 9 nm 0 W O mu 0 0 ¢ W W 0 u m a m w m 5 a». s 5 4 9.. 0 an :. coweomm m... an :. c.0323» a V... 239 DISCUSSION It has been well documented that the expression levels of GRK2 are altered in certain inflammatory disease conditions. For instance, GRK2 levels are significantly reduced in leukocytes from patients of active relapsing-remitting multiple sclerosis (MS) or with secondary progressive MS [3]. Peripheral Lymphocytes of patients with Alzheimers disease have an increased expression both mRNA and protein for GRK2 [16]. GRK expression have also been found to be altered in PBMCs of patients with rheumatoid arthiritis [5]. Although the significance of these changes is not clear particularly in these diseases, studies are just emerging on the pathophysiological role of GRK2 in these diseases in rodent models. For example, in an acute model of arthritis, GRK2 was shown to be a negative regulator of granulocyte chemotaxis to LTB4 (a potent Chemoattractant for granulocytes). Heterozygous GRK2 knockout mice showed an increased weight loss as well as development of arthritis was significantly enhanced in these mice [17]. It is, however, not clear what signals regulate the expression levels of GRK2. In a recent study, we demonstrated that TLR4 increases GRK2 levels in primary macrophages [15]. Other studies have demonstrated a similar role for TLR signaling in regulating GRK2 levels in immune cells. Although these studies proposed that the consequence of this increase is in limiting chemokine signaling, our studies presented here demonstrate that GRK2 regulates TLR4 signaling and LPS-induced cytokine/chemokine production and the consequent endotoxic shock. We show that GRK2 deficient mice show an enhanced liver and lung injury in response to LPS injection and are prone to LPS induced septic shock. This is not surprising in the light of the fact that other studies using heterozygous GRK2 mice 240 have shown similar results with other disease conditions. For instance, GRK2 +/- mice were found to be more susceptible for arthritis disease severity [l7]. GRK2 +/- animals also show an advanced onset of experimental autoimmune encephalomyelitis in association with an increased early cerebral infiltration of inflammatory cells [3]. Recent studies demonstrated that GRK2 +/- mice also show an increased susceptibility to neonatal hypoxic-ischemia brain damage [18]. While this manuscript was in preparation, another study showed the role of cell-type specific GRK2 in regulating hypoxia-ischemia brain damage [19]. Therefore, in general reduced levels of GRK2 increase the susceptibility to several disease conditions and we demonstrate here that myeloid cell GRK2 deficient mice are much more prone to LPS induced septic shock. The migration of neutrophils to the inflammatory sites although important for host defense, is also in part responsible for tissue damage observed in sepsis. MIPla, MIPlB, MCP1, Rantes and eotaxin are members of CC chemokine family which serve as major chemoattractants for neutrophils, mononuclear cells as well as eosinophils [20-23]. The secretary levels of all these chemokines were found to be enhanced in GRK2 deficient mice plasma. Out of these however, the levels of MIPla were found to be significantly increased both in plasma as well as cell culture supematants. Furthermore, MIPla was found to be specifically regulated by p105-ERK signaling since ERK inhibitor as well as IKK inhibitor inhibited its increased synthesis. MIPla is produced by several cell types including lymphocytes, monocytes/macrophages, mast cells, basophils, epithelial cells and fibroblasts and binds to CC chemokine receptor 1 (CCR1 and CCR5) to exert its biological actions. MIPla plays a crucial role in activation and chemotaxis of several populations of macrophages. It stimulates 241 the proliferation of mature tissue macrophages and also has been shown to induce the secretion of TNFa, IL-6 and IL-la from elicited peritoneal macrophages [24]. It is highly expressed in acute as well as chronic lung inflammation [25]. MIPla alpha has been previously shown to be responsible for LPS induced lung capillary leakage and early mortality in endotoxic shock [23]. We, in our studies also found a substantially increased lung capillary permeability as assessed by the measurement of total protein in the broncho-alveolar lavage fluid suggesting a role for increased MIPla in enhanced lung injury observed in our studies. Furthermore, there is an increased infiltration of inflammatory cells in the liver resulting in an increased liver damage. Hence, we propose MIPla to be an important factor responsible for inducing a substantially increased lung and liver injury in GRK2 deficient mice. Another important pro-inflammatory cytokine IL-12p40 is also found to be highly secreted in GRK2 deficient mouse plasma. It is a component of IL-12 cytokine and is an important mediator of cell mediated immunity [26]. Furthermore, it has been shown to play an important role in inflammatory diseases such as experimental autoimmune encephalitis [27]. IL-lO, an important anti-inflammatory cytokine was also found to be increased in GRK2 deficient mouse plasma. An increase in an anti-inflammatory cytokine should prevent from leading to endotoxemia in theory. However, studies have illustrated that although increase in IL-lO prevents an over—exuberant immune response, it can also lead to immunosuppression shifting the balance toward lethal consequencies. Since we observe an early lethality in LPS injected GRK2 deficient mice, it is suspected that the balance is shifted towards an increased inflammatory state leading to organ injury and mortality. 242 Our results demonstrate that the peritoneal macrophages from GRK2 deficient mice have an increased ERK activation in response to LPS treatment as compared to control mice. Inhibitor studies further support our findings that the increased ERK activity is responsible for the enhanced expression of certain pro-inflammatory mediators in GRK2 deficient peritoneal macrophages. The effect of GRK2 on ERK activation has also been shown by other investigators. In HEK 293 cells, increased GRK2 was found to cause reduced ERK activation in response to C-C chemokine ligand 2 (CCL2). On the other hand, splenocytes from GRK2+/- mice, were found to have an enhanced ERK activation in response to chemokines. These studies also showed that GRK2 and MEK are present in the same multi-molecular complex but unlike our studies, GRK2 did not affect MEK activity [28]. In another study, pro- inflarnmatory cytokine IL-lB was found to induce a 2—3 fold increase in the expression of GRK2 in primary astrocytes and a simultaneous decrease in CCL2-induced ERK1/2 activation whereas astrocytes from GRK2 +/- mice show an increase in ERKl/Z phosphorylation [29]. While these other studies have focused on chemokine receptor (a GPCR)-induced ERK activation, our studies address the role of GRK2 in TLR4 signaling, especially in primary macrophages. Thus although GRK2 has very important roles in GPCR signaling, our studies here demonstrate that GRK2 is equally important in regulating TLR4-induced ERK activation and the consequent cytokine/chemokine production. In this study we have determined that the mechanism by which GRK2 regulates TLR4-induced ERK activation lies at the level of NFKBl p105. In macrophages, NFKB] p105 lies upstream of MEK1/2 kinase and is present in a 243 complex with Tp12, a MAP3K. Our studies clearly show that LPS-induced MEK1/2 as well as NFKBI p105 phosphorylation are significantly enhanced in macrophages from GRK2 deficient mice. We believe that the mechanism lies at the level of NFKB] p105 for the following reasons: 1. LPS-induced Icha phosphorylation is not affected by GRK2 deficiency in macrophages. IKBOL is also phosphorylated by IKKB, the same enzyme that phosphorylates NFKB] p105. Thus if the regulation by GRK2 is at or above the level of IKKB, then one would expect that IKBa phosphorylation to also be regulated by GRK2. 2. Previous studies have shown that GRK2 and NFKB] p105 directly interact with each other. GRK2 and NFKB] p105 have been shown to interact in yeast two-hybrid assays. And the RH domain of GRK2 has been shown to directly interact with the Carboxy terminus of p105 in direct interaction assays. Previous studies, however, have also shown that NFKB] p105 is a poor substrate for GRK2, suggesting that the regulation might be phosphorylation independent. This is not entirely surprising given the role of RH domain of GRK2 in mediating phosphorylation-independent cell signaling, protentially as a scaffolding protein. In studies using Raw264.7 macrophages cell line, however, we did not find any functional significance of GRK2 in LPS-induced NFKBI p105-ERK pathway, using RNAi. In the present studies we find that in primary macrophages from genetically modified levels of GRK2, GRK2 does regulate LPS-induced NFKBI p105-ERK pathway. Thus the differences between the studies could be related to the differences in cell types (cell line v/s primary cells) and/or the method of decreasing GRK2 levels (RNAi v/s genetic). 244 GRK2 has previously been shown to regulate a number of signaling pathways not necessarily restricted to GPCR signaling. Even with in GPCRs, role of GRK2 has expanded considerably beyond its role in GPCR phosphorylation. Some of these signaling pathways are relevant to TLR4 signaling. For example, Liu et al., have demonstrated that Akt interacts with GRK2 and this interaction inhibits Akt activity, which then limits the activation of eNOS and the production of NO in sinusoidal endothelial cells leading to intrahepatic portal hypertension. Correspondingly, GRK2 deficient mice develop less severe portal hypertension after liver injury [30]. Even though TLR4 activation in primary macrophages induces Akt phosphorylation, we did not observe any difference in Akt phosphorylation between GRK2 deficient and control macrophages. One possibility for this difference is the different cell types examined. In another study, Peregrin et al. showed that GRK2 can interact with and phosphorylate p38 MAPK and this can lead to the inactivation of p38 MAPK. Furthermore, peritoneal macrophages from GRK2+/- mice were found to have an enhanced production of TNFa due to an enhanced activation of p38 [31]. In our studies, however, we did not observe enhanced activation of p38 in the GRK2 deficient macrophages compared to the control cells. The reason for this difference is not clear. In our studies, however, we demonstrate that inhibition of enhanced ERK activation significantly lowers the enhanced cytokine/chemokine levels to that of the wild type levels, suggesting a clear role for this signaling pathway in mediating MIPla, IL-loc, GCSF and MCP1. Our studies fiirther demonstrate that the NFKB] P105-mediated p50-NFKB pathway may also be equally important in mediating the enhanced secretion of IL-1 B and IL-6 in the GRK2 deficient macrophages because the 245 IKKB inhibitor blocked the production of these two cytokines even though they were not affected by the ERK inhibitor. It is not clear why we see a slightly different response in terms of the cytokines /chemokines which are secreted in plasma verses those in cell culture supematants. However, one can envision several possible reasons for this discrepancy: 1. A variety of cell types are involved in producing cytokines under in vivo conditions and the levels observed in plasma are an overall contribution of these various cell types as compared to only one cell type used in vitro in our studies. 2. LPS stimulation of TLR4 onto macrophages is a direct signaling mechanism leading to the expression of genes for cytokines / chemokines as compared to indirect signaling mechanisms due to cross talks involved under in vivo conditions. 2. We used only one type of macrophages i.e. thioglycollated elicited peritoneal macrophages under in vitro conditions. However, there are a variety of macrophages (tissue specific) which contribute to the effects seen under in vivo conditions. Several studies have shown that sepsis patients have increased circulatory levels of TNFa, MIPla, MIPlB, IL-6 and IL-10. Furthermore, in one study it was found that neutrophils from septic patients show a reduced chemotaxis due to a failure to induce an increase in tyrosine phosphorylation and actin polymerization in response to chemokines. These neutrophils were found to have high expression levels of GRK2 and GRK5. Moreover, neutrophils from healthy individuals treated with LPS and cytokines were also found to have an increased expression of GRK2 and GRK5. This suggested that the pro-inflammatory mediators produced during sepsis increase the expression of GRKs leading to desensitization of neutrophils to chemokines [8]. 246 However, our studies suggest that GRK2 is protective against sepsis in the initial phase since deletion of GRK2 in myeloid cells increases susceptibility to sepsis. In an already established septic condition however, the expression levels of GRK2 might increase in an effort to protect the system from the lethal consequences of sepsis. Reduced chemotaxis might be detrimental since neutrophils are required to help fight infection. The system, however, needs to maintain a balance and it is tempting to hypothesize that this might be mediated by increasing GRK2 levels under septic conditions. Obviously further studies are necessary to delineate these issues. In summary, mice with a deficiency of GRK2 in myeloid cells are more susceptible to LPS-mediated endotoxemia and this is associated with enhanced secretion of several cytokines/chemokines. We further demonstrate that the enhanced cytokine/chemokine in the GRK2 deficient mice is related to enhanced activation of the p105-ERK pathway in macrophages. Our results suggest that the increase in GRK2 levels observed in immune cells under septic conditions might limit the progression of sepsis. 247 REFERENCES Loudon, R. P., Perussia, B. and Benovic, J. L. (1996) Differentially regulated expression of the G-protein-coupled receptor kinases, betaARK and GRK6, during myelomonocytic cell development in vitro. Blood. 88, 4547—4557 De Blasi, A., Parruti, G. and Sallese, M. (1995) Regulation of G protein- coupled receptor kinase subtypes in activated T lymphocytes. Selective increase of beta-adrenergic receptor kinase 1 and 2. The Journal of clinical investigation. 95, 203-210 Vroon, A., Kavelaars, A., Limmroth, V., Lombardi, M. S., Goebel, M. U., Van Dam, A. M., Caron, M. G., Schedlowski, M. and Heijnen, C. J. (2005) G protein-coupled receptor kinase 2 in multiple sclerosis and experimental autoimmune encephalomyelitis. J Immunol. 174, 4400-4406 Giorelli, M., Livrea, P. and Trojano, M. (2004) Post-receptorial mechanisms underlie functional disregulation of beta2-adrenergic receptors in lymphocytes from Multiple Sclerosis patients. Journal of neuroimmunology. 155, 143-149 Lombardi, M. S., Kavelaars, A., Schedlowski, M., Bijlsma, J. W., Okihara, K. L., Van de Pol, M., Ochsmann, S., Pawlak, C., Schmidt, R. E. and Heijnen, C. J. (1999) Decreased expression and activity of G-protein-coupled receptor kinases in peripheral blood mononuclear cells of patients with rheumatoid arthritis. Faseb J. 13, 715—725 Vroon, A., Lombardi, M. S., Kavelaars, A. and Heijnen, C. J. (2003) Changes in the G-protein-coupled receptor desensitization machinery during relapsing- progressive experimental allergic encephalomyelitis. Journal of neuroimmunology. 137, 79-86 Lombardi, M. S., Kavelaars, A., Cobelens, P. M., Schmidt, R. E., Schedlowski, M. and Heijnen, C. J. (2001) Adjuvant arthritis induces down-regulation of G protein-coupled receptor kinases in the immune system. J Immunol. 166, 163 5- 1640 Arraes, S. M., Freitas, M. 8., da Silva, S. V., de Paula Neto, H. A., Alves-Filho, J. C., Auxiliadora Martins, M., Basile-Filho, A., Tavares-Murta, B. M., Barja- Fidalgo, C. and Cunha, F. Q. (2006) Impaired neutrophil chemotaxis in sepsis associates with GRK expression and inhibition of actin assembly and tyrosine phosphorylation. Blood. 108, 2906-2913 248 10 ll 12 13 14 15 16 17 O'Neill, L. A. and Bowie, A. G. (2007) The family of five: TIR-domain- containing adaptors in Toll-like receptor signalling. Nature reviews. 7, 353-364 Clausen, B. E., Burkhardt, C., Reith, W., Renkawitz, R. and Forster, I. ( 1999) Conditional gene targeting in macrophages and granulocytes using LysMcre mice. Transgenic research. 8, 265-277 Matkovich, S. J ., Diwan, A., Klanke, J. L., Hammer, D. J ., Marreez, Y., Odley, A. M., Brunskill, E. W., Koch, W. J., Schwartz, R. J. and Dom, G. W., 2nd. (2006) Cardiac-specific ablation of G-protein receptor kinase 2 redefines its roles in heart development and beta-adrenergic signaling. Circulation research. 99, 996-1003 Appledom, D. M., Patial, S., McBride, A., Godbehere, S., Van Rooijen, N., Parameswaran, N. and Amalfitano, A. (2008) Adenovirus vector-induced innate inflammatory mediators, MAPK signaling, as well as adaptive immune responses are dependent upon both TLR2 and TLR9 in vivo. J Immunol. 181, 2134-2144 Patial, S., Luo, J., Porter, K. J ., Benovic, J. L. and Parameswaran, N. (2009) G- protein coupled receptor kinases mediate TNFalpha-induced NFkappaB signaling via direct interaction with and phosphorylation of IkappaBalpha. The Biochemical journal Moosbauer, C., Morgenstern, E., Cuvelier, S. L., Manukyan, D., Bidzhekov, K., Albrecht, S., Lohse, R, Patel, K. D. and Engelrnann, B. (2007) Eosinophils are a major intravascular location for tissue factor storage and exposure. Blood. 109, 995-1002 Loniewski, K., Shi, Y., Pestka, J. and Parameswaran, N. (2008) Toll-like receptors differentially regulate GPCR kinases and arrestins in primary macrophages. Molecular immunology. 45, 2312-2322 Leosco, D., Fortunato, F., Rengo, G., Iaccarino, G., Sanzari, E., Golino, L., Zincarelli, C., Canonico, V., Marchese, M., Koch, W. J. and Rengo, F. (2007) Lymphocyte G-protein-coupled receptor kinase-2 is upregulated in patients with Alzheimer's disease. Neuroscience letters. 415, 27 9-282 Tarrant, T. K., Rampersad, R. R., Esserrnan, D., Rothlein, L. R., Liu, P., Premont, R. T., Lefl