Comprehensive Physiology Wiley Online Library

GPCRs, G Proteins, and Their Impact on β‐cell Function

Full Article on Wiley Online Library



Abstract

Glucose‐induced (physiological) insulin secretion from the islet β‐cell involves interplay between cationic (i.e., changes in intracellular calcium) and metabolic (i.e., generation of hydrophobic and hydrophilic second messengers) events. A large body of evidence affirms support for novel regulation, by G proteins, of specific intracellular signaling events, including actin cytoskeletal remodeling, transport of insulin‐containing granules to the plasma membrane for fusion, and secretion of insulin into the circulation. This article highlights the following aspects of GPCR‐G protein biology of the islet. First, it overviews our current understanding of the identity of a wide variety of G protein regulators and their modulatory roles in GPCR‐G protein‐effector coupling, which is requisite for optimal β‐cell function under physiological conditions. Second, it describes evidence in support of novel, noncanonical, GPCR‐independent mechanisms of activation of G proteins in the islet. Third, it highlights the evidence indicating that abnormalities in G protein function lead to islet β‐cell dysregulation and demise under the duress of metabolic stress and diabetes. Fourth, it summarizes observations of potential beneficial effects of GPCR agonists in preventing/halting metabolic defects in the islet β‐cell under various pathological conditions (e.g., metabolic stress and inflammation). Lastly, it identifies knowledge gaps and potential avenues for future research in this evolving field of translational islet biology. Published 2020. Compr Physiol 10:453‐490, 2020.

Keywords: hormone receptors; cell physiology; intracellular signaling; cell physiology; diabetes; endocrinology; metabolism

Comprehensive Physiology offers downloadable PowerPoint presentations of figures for non-profit, educational use, provided the content is not modified and full credit is given to the author and publication.

Download a PowerPoint presentation of all images


Figure 1. Figure 1. Schematic representation of GPCR‐G protein‐effector coupling. Binding of a ligand to an appropriate GPCR leads to activation of a heterotrimeric G protein via GDP/GTP exchange on the α‐subunit. The GTP‐bound form of the α‐subunit (Gα.GTP) is functionally active and dissociates from the βγ subunit complex for activation of variety of effector proteins. The GSα.GTP and Giα.GTP promote activation and inactivation of adenylyl cyclases, respectively, leading to either increased or decreased generation of cAMP levels intracellularly. Gqα.GTP has been implicated in activation of PLases leading to the generation of biologically active hydrophobic (e.g., DAG) and hydrophilic (IP3) second messengers of insulin secretion. DAG and IP3 regulate protein kinase C and mobilization of intracellular calcium from the intracellular pools, respectively. Activation of G12α.GTP results in the regulation of downstream signaling pathways, including activation of Rho subfamily of G proteins, which have been shown to play critical modulatory roles in vesicular transport and actin cytoskeletal remodeling. Lastly, the βγ complex exerts regulatory effects on a wide variety of signaling pathways, including activation of PLases and adenylyl cyclases.
Figure 2. Figure 2. Activation‐deactivation of heterotrimeric G proteins. In its inactive conformation, a heterotrimeric G protein remains as α.GDP/βγ complex. Following activation by a GPCR (Figure 1), the α.GTP dissociates from βγ complex to precisely control their respective effector proteins. Exchange of GDP for GTP is mediated via a variety of GEFs. Likewise, the hydrolysis of GTP bound to the Gα subunit is regulated by a number of regulatory proteins and factors (see text for additional details). GEF, GTP/GDP exchange factor; GAP, GTPase‐activating protein; R, receptor.
Figure 3. Figure 3. Regulatory roles of small G proteins in glucose‐stimulated insulin secretion. A large body of experimental evidence supports the key roles of small G proteins in the stimulus‐secretion coupling of GSIS from the pancreatic β‐cell. Shown here are examples of some of these G proteins and their regulatory roles in mediating actin cytoskeletal remodeling, priming/docking, and fusion of insulin‐laden secretory granules with the plasma membrane for exocytotic secretion of insulin. Published evidence suggests sequential activation of these G proteins in the cascade of events leading to GSIS. For example, it was demonstrated that GSIS involves successive activation of Arf6 (∼1 min), Cdc42 (∼3 min), and Rac1 (∼15 min) 90. In addition to these, several groups of proteins have been identified and characterized in the islet β‐cell, which are essential for recognition and docking of the secretory vesicles on the plasma membrane 127,298.
Figure 4. Figure 4. Activation‐deactivation of small G proteins. Small G proteins (e.g., Cdc42 and Rac1) undergo activation (GTP‐bound) and deactivation (GDP‐bound) cycles. Inactive (GDP‐bound) G proteins are converted to their active (GTP‐bound) counterparts by GEFs. Following transmission of signals requisite for the effector activation, the active form of a candidate G protein returns to its inactive form; such a transition is mediated by the GTPase activity intrinsic to the G protein. In addition, a class of regulatory proteins, referred to as the GTPase‐activating proteins (GAPs), has been shown to accelerate the GTPase activity. Lastly, the GDP‐dissociation inhibitors (GDIs) play key roles in maintaining the candidate G proteins in their GDP‐bound (inactive) configuration. GDIs also contribute toward the targeting of G proteins to appropriate subcellular compartments (see text for additional details). GEFs, GTP/GDP exchange factors; GDI, GDP‐dissociation inhibitor; GAP, GTPase‐activating protein.
Figure 5. Figure 5. Examples of regulators of small G protein functions in the islet β‐cell. A depiction of various regulatory factors of G protein function which are identified in the pancreatic β‐cell. In addition to GEFs, GAPs, and GDIs, several other G protein regulatory proteins/factors (listed under “others”) have been identified and studied in the pancreatic β‐cell. Their modulatory roles are described in the narrative.
Figure 6. Figure 6. Proposed model for NDPK‐mediated, GPCR‐independent (noncanonical) activation of heterotrimeric G proteins. Based on the available evidence we propose a mechanism for an alternate, GPCR‐independent (noncanonical) activation of heterotrimeric G proteins in the pancreatic β‐cell by glucose. Trimeric G proteins remain inactive as a α.GDP/βγ complex. NDPK/histidine kinases catalyze phosphorylation of the β‐subunit at a histidine residue (via a phosphoramidate linkage). This phosphate, in turn, is relayed to Gα.GDP to yield its GTP‐bound (active) conformation. This is analogous to the classical ping‐pong mechanism of high‐energy phosphate transfer mediated by NDPK family of enzymes. Upon activation, Gα.GTP dissociates from the βγ complex; both of these entities regulate their respective effector proteins, leading to activation or inactivation of downstream signaling pathways (e.g., activation/inactivation of adenylyl cyclase for the generation or suppression of cAMP production). Following this, the GTPase activity, which is intrinsic to the Gα subunit, hydrolyzes the GTP bound to the α‐subunit to GDP (inactive conformation), thus facilitating the complexation of the βγ subunit with Gα.GDP. G, guanosine group.
Figure 7. Figure 7. Examples of noncanonical modulators of heterotrimeric G proteins. Depicted herein are potential alternate (noncanonical) regulators of heterotrimeric G proteins. Binding of ligands to their cognate GPCRs leads to activation of G proteins via GTP/GDP exchange. Recent studies have identified several non‐GPCR‐dependent regulatory mechanisms for GTP/GDP exchange including those involving NDPKs. In addition to these, several other modes of noncanonical activation of trimeric G proteins have been described (see text). These include Ric‐8A and GEMs (e.g., GIV/Girdin). The GEM class of proteins has been shown to regulate signaling pathways downstream to cell surface receptors. Furthermore, a novel class of regulatory proteins, namely activators of G protein signaling (AGS), has also been identified and studied well in other cell types, but not in the context of islet β‐cell function. Altogether, these regulators are involved in GTP/GDP exchange culminating in the formation of Gα.GTP (active) for effector activation. In a manner akin to the canonically activated trimeric G proteins, the βγ subunits regulate various effectors. Following effector activation, the active form of Gα is converted to its inactive GDP‐bound conformation through the intermediacy of intrinsic GTPase. In addition to this, a novel class of regulators of G protein signaling (RGS) has been identified in many cell types, including the islet β‐cell. The RGS proteins serve as GAPs in accelerating the GTPase activity, thereby terminating the GPCR activation signals. The identity and functional roles of various RGS proteins are highlighted in the text and in Table 3.
Figure 8. Figure 8. Posttranslational modifications of small G proteins and γ‐subunits of trimeric G proteins. (A) Mevalonic acid is the precursor for the biosynthesis of FPP and GGPP. FTase catalyzes the farnesylation of small G proteins (e.g., Ras), certain γ‐subunits of trimeric G proteins, and nuclear lamins A and B. GGTase‐I mediates the geranylgeranylation Rho subfamily of G proteins including Rho, Cdc42, Rac1, and Rap1. GGTase‐II catalyzes geranylgeranylation Rab subfamily of G proteins. (B) First, FTase or GGTase mediates incorporation of prenyl pyrophosphates into the C‐terminal cysteine of G proteins. Second, three amino acids after the farnesylated or geranylgeranylated cysteine are removed by a protease (Rce1). Third, isoprenylcysteine carboxyl methyltransferase (ICMT) catalyzes methylation of the carboxylate group of the prenylated cysteine using S‐adenosyl methionine as the methyl donor 131,137. Not shown in the schematic are additional posttranslational modification steps (e.g., palmitoylation) at a cysteine residue, which is upstream to the prenylated cysteines of G proteins (e.g., Ras and Rac1). This figure is reproduced with permission from Elsevier 137.
Figure 9. Figure 9. Our working model for mechanisms underlying glucose‐induced G protein‐mediated insulin secretion. Based on the data accrued in multiple investigations, we propose a model for NDPK/HK signaling step in the stimulus‐secretion coupling of GSIS in the islet β‐cell. Glucose metabolism leads to activation of PLases (e.g., PLase‐C and ‐D), resulting in the generation of biologically active hydrophobic as well as hydrophilic lipid messengers, which, in turn, activate protein histidine kinases. Furthermore, these biologically active lipids have been shown to regulate GTP‐binding and GTPase activities of G proteins in various subcellular fractions derived from the islet. In addition, these second messengers have been shown to promote translocation and membrane association of small G proteins, such as Rac1. Furthermore, activation of protein histidine phosphorylation, by NADPKs, leads to increase in GTP/GDP and GTP channeling to G proteins for their activation. Lastly, findings from our earlier studies suggested novel roles for protein histidine phosphorylation in the regulation of enzymes of glucose metabolism, such as mitochondrial succinyl‐coA synthase 121. Based on this experimental evidence, we propose that protein histidine phosphorylation contributes to activation of multiple signaling events in the cascade of events leading to GSIS.
Figure 10. Figure 10. Proposed mechanisms for ROS‐dependent regulation of islet β‐cell function in normal health and under the duress of metabolic stress. Several lines of evidence implicate positive and negative modulatory roles for ROS in islet function. NADPH oxidases (Noxs; e.g., Nox1, Nox2, and Nox4) represent the key sources for ROS generation in the islet β‐cell. Under physiological conditions, a tonic increase in the intracellular ROS leads to activation of Raf‐Ras signaling pathway, which is upstream to ERK1/2 and Rac1 activation, leading to GSIS 149. It has been suggested that intracellularly generated ROS could regulate a variety of cellular events including actin cytoskeletal remodeling 276 and proliferation 171. Therefore, a tonic increase in ROS might promote cytoskeletal remodeling, insulin secretion, and cell proliferation events in the islet β‐cell. On the other hand, excessive ROS generation, mediated via activation of various Nox isoforms under the duress of metabolic stress and exposure to pro‐inflammatory cytokines, manifests in increased intracellular oxidative stress resulting in the constitutive activation of Rac1 and subsequent activation of downstream stress kinases (JNK1/2, p38, and p53), resulting in cell demise via accelerated apoptosis 131,250.
Figure 11. Figure 11. Proposed cross talk between various signaling proteins in the GPCR‐G protein‐effector signalome culminating in physiological insulin secretion. GPCR agonists and fatty acids bind to their cognate receptors on the plasma membrane, leading to the activation of respective heterotrimeric G proteins (Figure 1). Exposure of the islet β‐cell to stimulatory glucose leads to its metabolism and activation of signaling pathways, including activation of NDPKs (and other signaling proteins; Figures 7 and 9), culminating in the activation of trimeric G proteins in a noncanonical mode. Activation of GPCR‐G protein signaling pathway results in the generation of second messengers of insulin secretion, including cAMP, IP3, DAG, and biologically active phospholipids (e.g., lyso‐phospholipids). These, in turn, promote activation of small G proteins belonging to Rho and Rab subfamilies, leading to cytoskeletal remodeling, vesicular transport, and fusion of secretory granules with the plasma membrane and secretion of insulin via exocytosis. As discussed in this article, activation of G proteins (heterotrimeric and small G proteins) is controlled precisely by a wide array of regulatory factors, including GEFs, GAPs, GDIs, and others (e.g., AGS proteins, GEMs, and RGS proteins). Furthermore, functional activation of G proteins and their regulators appears to be mediated precisely via posttranslational modifications, including phosphorylation, prenylation, carboxylmethylation, palmitoylation, ubiquitination, and SUMOylation. Not depicted in this figure are potential possibilities for defects in the functional activation of these regulators (e.g., mutations) under the duress of metabolic stress that could result in abnormalities in cross talk within this signalome, resulting in the apoptotic demise of the islet β‐cell, and the onset of diabetes.


Figure 1. Schematic representation of GPCR‐G protein‐effector coupling. Binding of a ligand to an appropriate GPCR leads to activation of a heterotrimeric G protein via GDP/GTP exchange on the α‐subunit. The GTP‐bound form of the α‐subunit (Gα.GTP) is functionally active and dissociates from the βγ subunit complex for activation of variety of effector proteins. The GSα.GTP and Giα.GTP promote activation and inactivation of adenylyl cyclases, respectively, leading to either increased or decreased generation of cAMP levels intracellularly. Gqα.GTP has been implicated in activation of PLases leading to the generation of biologically active hydrophobic (e.g., DAG) and hydrophilic (IP3) second messengers of insulin secretion. DAG and IP3 regulate protein kinase C and mobilization of intracellular calcium from the intracellular pools, respectively. Activation of G12α.GTP results in the regulation of downstream signaling pathways, including activation of Rho subfamily of G proteins, which have been shown to play critical modulatory roles in vesicular transport and actin cytoskeletal remodeling. Lastly, the βγ complex exerts regulatory effects on a wide variety of signaling pathways, including activation of PLases and adenylyl cyclases.


Figure 2. Activation‐deactivation of heterotrimeric G proteins. In its inactive conformation, a heterotrimeric G protein remains as α.GDP/βγ complex. Following activation by a GPCR (Figure 1), the α.GTP dissociates from βγ complex to precisely control their respective effector proteins. Exchange of GDP for GTP is mediated via a variety of GEFs. Likewise, the hydrolysis of GTP bound to the Gα subunit is regulated by a number of regulatory proteins and factors (see text for additional details). GEF, GTP/GDP exchange factor; GAP, GTPase‐activating protein; R, receptor.


Figure 3. Regulatory roles of small G proteins in glucose‐stimulated insulin secretion. A large body of experimental evidence supports the key roles of small G proteins in the stimulus‐secretion coupling of GSIS from the pancreatic β‐cell. Shown here are examples of some of these G proteins and their regulatory roles in mediating actin cytoskeletal remodeling, priming/docking, and fusion of insulin‐laden secretory granules with the plasma membrane for exocytotic secretion of insulin. Published evidence suggests sequential activation of these G proteins in the cascade of events leading to GSIS. For example, it was demonstrated that GSIS involves successive activation of Arf6 (∼1 min), Cdc42 (∼3 min), and Rac1 (∼15 min) 90. In addition to these, several groups of proteins have been identified and characterized in the islet β‐cell, which are essential for recognition and docking of the secretory vesicles on the plasma membrane 127,298.


Figure 4. Activation‐deactivation of small G proteins. Small G proteins (e.g., Cdc42 and Rac1) undergo activation (GTP‐bound) and deactivation (GDP‐bound) cycles. Inactive (GDP‐bound) G proteins are converted to their active (GTP‐bound) counterparts by GEFs. Following transmission of signals requisite for the effector activation, the active form of a candidate G protein returns to its inactive form; such a transition is mediated by the GTPase activity intrinsic to the G protein. In addition, a class of regulatory proteins, referred to as the GTPase‐activating proteins (GAPs), has been shown to accelerate the GTPase activity. Lastly, the GDP‐dissociation inhibitors (GDIs) play key roles in maintaining the candidate G proteins in their GDP‐bound (inactive) configuration. GDIs also contribute toward the targeting of G proteins to appropriate subcellular compartments (see text for additional details). GEFs, GTP/GDP exchange factors; GDI, GDP‐dissociation inhibitor; GAP, GTPase‐activating protein.


Figure 5. Examples of regulators of small G protein functions in the islet β‐cell. A depiction of various regulatory factors of G protein function which are identified in the pancreatic β‐cell. In addition to GEFs, GAPs, and GDIs, several other G protein regulatory proteins/factors (listed under “others”) have been identified and studied in the pancreatic β‐cell. Their modulatory roles are described in the narrative.


Figure 6. Proposed model for NDPK‐mediated, GPCR‐independent (noncanonical) activation of heterotrimeric G proteins. Based on the available evidence we propose a mechanism for an alternate, GPCR‐independent (noncanonical) activation of heterotrimeric G proteins in the pancreatic β‐cell by glucose. Trimeric G proteins remain inactive as a α.GDP/βγ complex. NDPK/histidine kinases catalyze phosphorylation of the β‐subunit at a histidine residue (via a phosphoramidate linkage). This phosphate, in turn, is relayed to Gα.GDP to yield its GTP‐bound (active) conformation. This is analogous to the classical ping‐pong mechanism of high‐energy phosphate transfer mediated by NDPK family of enzymes. Upon activation, Gα.GTP dissociates from the βγ complex; both of these entities regulate their respective effector proteins, leading to activation or inactivation of downstream signaling pathways (e.g., activation/inactivation of adenylyl cyclase for the generation or suppression of cAMP production). Following this, the GTPase activity, which is intrinsic to the Gα subunit, hydrolyzes the GTP bound to the α‐subunit to GDP (inactive conformation), thus facilitating the complexation of the βγ subunit with Gα.GDP. G, guanosine group.


Figure 7. Examples of noncanonical modulators of heterotrimeric G proteins. Depicted herein are potential alternate (noncanonical) regulators of heterotrimeric G proteins. Binding of ligands to their cognate GPCRs leads to activation of G proteins via GTP/GDP exchange. Recent studies have identified several non‐GPCR‐dependent regulatory mechanisms for GTP/GDP exchange including those involving NDPKs. In addition to these, several other modes of noncanonical activation of trimeric G proteins have been described (see text). These include Ric‐8A and GEMs (e.g., GIV/Girdin). The GEM class of proteins has been shown to regulate signaling pathways downstream to cell surface receptors. Furthermore, a novel class of regulatory proteins, namely activators of G protein signaling (AGS), has also been identified and studied well in other cell types, but not in the context of islet β‐cell function. Altogether, these regulators are involved in GTP/GDP exchange culminating in the formation of Gα.GTP (active) for effector activation. In a manner akin to the canonically activated trimeric G proteins, the βγ subunits regulate various effectors. Following effector activation, the active form of Gα is converted to its inactive GDP‐bound conformation through the intermediacy of intrinsic GTPase. In addition to this, a novel class of regulators of G protein signaling (RGS) has been identified in many cell types, including the islet β‐cell. The RGS proteins serve as GAPs in accelerating the GTPase activity, thereby terminating the GPCR activation signals. The identity and functional roles of various RGS proteins are highlighted in the text and in Table 3.


Figure 8. Posttranslational modifications of small G proteins and γ‐subunits of trimeric G proteins. (A) Mevalonic acid is the precursor for the biosynthesis of FPP and GGPP. FTase catalyzes the farnesylation of small G proteins (e.g., Ras), certain γ‐subunits of trimeric G proteins, and nuclear lamins A and B. GGTase‐I mediates the geranylgeranylation Rho subfamily of G proteins including Rho, Cdc42, Rac1, and Rap1. GGTase‐II catalyzes geranylgeranylation Rab subfamily of G proteins. (B) First, FTase or GGTase mediates incorporation of prenyl pyrophosphates into the C‐terminal cysteine of G proteins. Second, three amino acids after the farnesylated or geranylgeranylated cysteine are removed by a protease (Rce1). Third, isoprenylcysteine carboxyl methyltransferase (ICMT) catalyzes methylation of the carboxylate group of the prenylated cysteine using S‐adenosyl methionine as the methyl donor 131,137. Not shown in the schematic are additional posttranslational modification steps (e.g., palmitoylation) at a cysteine residue, which is upstream to the prenylated cysteines of G proteins (e.g., Ras and Rac1). This figure is reproduced with permission from Elsevier 137.


Figure 9. Our working model for mechanisms underlying glucose‐induced G protein‐mediated insulin secretion. Based on the data accrued in multiple investigations, we propose a model for NDPK/HK signaling step in the stimulus‐secretion coupling of GSIS in the islet β‐cell. Glucose metabolism leads to activation of PLases (e.g., PLase‐C and ‐D), resulting in the generation of biologically active hydrophobic as well as hydrophilic lipid messengers, which, in turn, activate protein histidine kinases. Furthermore, these biologically active lipids have been shown to regulate GTP‐binding and GTPase activities of G proteins in various subcellular fractions derived from the islet. In addition, these second messengers have been shown to promote translocation and membrane association of small G proteins, such as Rac1. Furthermore, activation of protein histidine phosphorylation, by NADPKs, leads to increase in GTP/GDP and GTP channeling to G proteins for their activation. Lastly, findings from our earlier studies suggested novel roles for protein histidine phosphorylation in the regulation of enzymes of glucose metabolism, such as mitochondrial succinyl‐coA synthase 121. Based on this experimental evidence, we propose that protein histidine phosphorylation contributes to activation of multiple signaling events in the cascade of events leading to GSIS.


Figure 10. Proposed mechanisms for ROS‐dependent regulation of islet β‐cell function in normal health and under the duress of metabolic stress. Several lines of evidence implicate positive and negative modulatory roles for ROS in islet function. NADPH oxidases (Noxs; e.g., Nox1, Nox2, and Nox4) represent the key sources for ROS generation in the islet β‐cell. Under physiological conditions, a tonic increase in the intracellular ROS leads to activation of Raf‐Ras signaling pathway, which is upstream to ERK1/2 and Rac1 activation, leading to GSIS 149. It has been suggested that intracellularly generated ROS could regulate a variety of cellular events including actin cytoskeletal remodeling 276 and proliferation 171. Therefore, a tonic increase in ROS might promote cytoskeletal remodeling, insulin secretion, and cell proliferation events in the islet β‐cell. On the other hand, excessive ROS generation, mediated via activation of various Nox isoforms under the duress of metabolic stress and exposure to pro‐inflammatory cytokines, manifests in increased intracellular oxidative stress resulting in the constitutive activation of Rac1 and subsequent activation of downstream stress kinases (JNK1/2, p38, and p53), resulting in cell demise via accelerated apoptosis 131,250.


Figure 11. Proposed cross talk between various signaling proteins in the GPCR‐G protein‐effector signalome culminating in physiological insulin secretion. GPCR agonists and fatty acids bind to their cognate receptors on the plasma membrane, leading to the activation of respective heterotrimeric G proteins (Figure 1). Exposure of the islet β‐cell to stimulatory glucose leads to its metabolism and activation of signaling pathways, including activation of NDPKs (and other signaling proteins; Figures 7 and 9), culminating in the activation of trimeric G proteins in a noncanonical mode. Activation of GPCR‐G protein signaling pathway results in the generation of second messengers of insulin secretion, including cAMP, IP3, DAG, and biologically active phospholipids (e.g., lyso‐phospholipids). These, in turn, promote activation of small G proteins belonging to Rho and Rab subfamilies, leading to cytoskeletal remodeling, vesicular transport, and fusion of secretory granules with the plasma membrane and secretion of insulin via exocytosis. As discussed in this article, activation of G proteins (heterotrimeric and small G proteins) is controlled precisely by a wide array of regulatory factors, including GEFs, GAPs, GDIs, and others (e.g., AGS proteins, GEMs, and RGS proteins). Furthermore, functional activation of G proteins and their regulators appears to be mediated precisely via posttranslational modifications, including phosphorylation, prenylation, carboxylmethylation, palmitoylation, ubiquitination, and SUMOylation. Not depicted in this figure are potential possibilities for defects in the functional activation of these regulators (e.g., mutations) under the duress of metabolic stress that could result in abnormalities in cross talk within this signalome, resulting in the apoptotic demise of the islet β‐cell, and the onset of diabetes.
References
 1.AbdAlla S, Lother H, el Missiry A, Langer A, Sergeev P, el Faramawy Y, Quitterer U. Angiotensin II AT2 receptor oligomers mediate G protein dysfunction in an animal model of Alzheimer disease. J Biol Chem 284: 6554‐6565, 2009.
 2.AbdAlla S, Lother H, el Missiry A, Sergeev P, Langer A, el Faramawy Y, Quitterer U. Dominant negative AT2 receptor oligomers induce G protein arrest and symptoms of neurodegeneration. J Biol Chem 284: 6566‐6574, 2009.
 3.Abu‐Taha IH, Heijman J, Feng Y, Vettel C, Dobrev D, Wieland T. Regulation of heterotrimeric G protein signaling by NDPK/NME proteins and caveolins: An update. Lab Invest 98: 190‐197, 2018.
 4.Acharya JD, Ghaskadbi SS. Islets and their antioxidant defense. Islets 2: 225‐235, 2010.
 5.Adra CN, Iyengar AR, Syed FA, Kanaan IN, Rilo HL, Yu W, Kheraj R, Lin SR, Horiuchi T, Khan S, Weremowicz S, Lim B, Morton CC, Higgs DR. Human ARHGDIG, a GDP‐dissociation inhibitor for Rho proteins: Genomic structure, sequence, expression analysis, and mapping to chromosome 16p13.3. Genomics 53: 104‐109, 1998.
 6.Ahren B. Islet G protein‐coupled receptors as potential targets for treatment of type 2 diabetes. Nat Rev Drug Discov 8: 369‐385, 2009.
 7.Aizawa T, Komatsu M. Rab27a: A new face in beta cell metabolism‐secretion coupling. J Clin Invest 115: 227‐230, 2005.
 8.Alexandrov K, Horiuchi H, Steele‐Mortimer O, Seabra MC, Zerial M. Rab escort protein‐1 is a multifunctional protein that accompanies newly prenylated rab proteins to their target membranes. EMBO J 13: 5262‐5273, 1994.
 9.Alory C, Balch WE. Molecular basis for Rab prenylation. J Cell Biol 150: 89‐103, 2000.
 10.Alqinyah M, Hooks SB. Regulating the regulators: Epigenetic, transcriptional, and post‐translational regulation of RGS proteins. Cell Signal 42: 77‐87, 2018.
 11.Alves ID, Lecomte S. Study of G protein coupled receptor signaling in membrane environment by plasmon waveguide resonance. Acc Chem Res 52: 1059‐1067, 2019.
 12.Amin RH, Chen HQ, Veluthakal R, Silver RB, Li J, Li G, Kowluru A. Mastoparan‐induced insulin secretion from insulin‐secreting betaTC3 and INS‐1 cells: Evidence for its regulation by Rho subfamily of G proteins. Endocrinology 144: 4508‐4518, 2003.
 13.Amisten S, Salehi A, Rorsman P, Jones PM, Persaud SJ. An atlas and functional analysis of G protein coupled receptors in human islets of Langerhans. Pharmacol Ther 139: 359‐391, 2013.
 14.Anvari E, Wikstrom P, Walum E, Welsh N. The novel NADPH oxidase 4 inhibitor GLX351322 counteracts glucose intolerance in high‐fat diet‐treated C57BL/6 mice. Free Radic Res 49: 1308‐1318, 2015.
 15.Arava Y, Seger R, Walker MD. GRFbeta, a novel regulator of calcium signaling, is expressed in pancreatic beta cells and brain. J Biol Chem 274: 24449‐24452, 1999.
 16.Arora DK, Syed I, Machhadieh B, McKenna CE, Kowluru A. Rab‐geranylgeranyl transferase regulates glucose‐stimulated insulin secretion from pancreatic beta cells. Islets 4: 354‐358, 2012.
 17.Asahara S, Shibutani Y, Teruyama K, Inoue HY, Kawada Y, Etoh H, Matsuda T, Kimura‐Koyanagi M, Hashimoto N, Sakahara M, Fujimoto W, Takahashi H, Ueda S, Hosooka T, Satoh T, Inoue H, Matsumoto M, Aiba A, Kasuga M, Kido Y. Ras‐related C3 botulinum toxin substrate 1 (RAC1) regulates glucose‐stimulated insulin secretion via modulation of F‐actin. Diabetologia 56: 1088‐1097, 2013.
 18.Attwood PV, Wieland T. Nucleoside diphosphate kinase as protein histidine kinase. Naunyn Schmiedebergs Arch Pharmacol 388: 153‐160, 2015.
 19.Baidwan S, Chekuri A, Hynds DL, Kowluru A. Glucotoxicity promotes aberrant activation and mislocalization of Ras‐related C3 botulinum toxin substrate 1 [Rac1] and metabolic dysfunction in pancreatic islet beta‐cells: Reversal of such metabolic defects by metformin. Apoptosis 22: 1380‐1393, 2017.
 20.Blumer JB, Lanier SM. Activators of G protein signaling exhibit broad functionality and define a distinct core signaling triad. Mol Pharmacol 85: 388‐396, 2014.
 21.Boissan M, Dabernat S, Peuchant E, Schlattner U, Lascu I, Lacombe ML. The mammalian Nm23/NDPK family: From metastasis control to cilia movement. Mol Cell Biochem 329: 51‐62, 2009.
 22.Boularan C, Kehrl JH. Implications of non‐canonical G protein signaling for the immune system. Cell Signal 26: 1269‐1282, 2014.
 23.Bourne HR. How receptors talk to trimeric G proteins. Curr Opin Cell Biol 9: 134‐142, 1997.
 24.Bouzakri K, Ribaux P, Tomas A, Parnaud G, Rickenbach K, Halban PA. Rab GTPase‐activating protein AS160 is a major downstream effector of protein kinase B/Akt signaling in pancreatic beta‐cells. Diabetes 57: 1195‐1204, 2008.
 25.Brandt DT, Grosse R. Get to grips: Steering local actin dynamics with IQGAPs. EMBO Rep 8: 1019‐1023, 2007.
 26.Brown AM, Birnbaumer L. Direct G protein gating of ion channels. Am J Physiol 254: H401‐H410, 1988.
 27.Brunk I, Holtje M, von Jagow B, Winter S, Sternberg J, Blex C, Pahner I, Ahnert‐Hilger G. Regulation of vesicular monoamine and glutamate transporters by vesicle‐associated trimeric G proteins: New jobs for long‐known signal transduction molecules. Handb Exp Pharmacol 175: 305‐325, 2006.
 28.Burhani MD, Rasenick MM. Fish oil and depression: The skinny on fats. J Integr Neurosci 16: S115‐S124, 2017.
 29.Bustelo XR. Vav family exchange factors: An integrated regulatory and functional view. Small GTPases 5: 9, 2014.
 30.Buteau J, El‐Assaad W, Rhodes CJ, Rosenberg L, Joly E, Prentki M. Glucagon‐like peptide‐1 prevents beta cell glucolipotoxicity. Diabetologia 47: 806‐815, 2004.
 31.Calizo RC, Scarlata S. A role for G proteins in directing G protein‐coupled receptor‐caveolae localization. Biochemistry 51: 9513‐9523, 2012.
 32.Campbell AP, Smrcka AV. Targeting G protein‐coupled receptor signalling by blocking G proteins. Nat Rev Drug Discov 17: 789‐803, 2018.
 33.Campden R, Audet N, Hebert TE. Nuclear G protein signaling: New tricks for old dogs. J Cardiovasc Pharmacol 65: 110‐122, 2015.
 34.Castillo‐Lluva S, Tatham MH, Jones RC, Jaffray EG, Edmondson RD, Hay RT, Malliri A. SUMOylation of the GTPase Rac1 is required for optimal cell migration. Nat Cell Biol 12: 1078‐1085, 2010.
 35.Cazares VA, Subramani A, Saldate JJ, Hoerauf W, Stuenkel EL. Distinct actions of Rab3 and Rab27 GTPases on late stages of exocytosis of insulin. Traffic 15: 997‐1015, 2014.
 36.Cerione RA. The experiences of a biochemist in the evolving world of G protein‐dependent signaling. Cell Signal 41: 2‐8, 2018.
 37.Cernohorska M, Sulimenko V, Hajkova Z, Sulimenko T, Sladkova V, Vinopal S, Draberova E, Draber P. GIT1/betaPIX signaling proteins and PAK1 kinase regulate microtubule nucleation. Biochim Biophys Acta 1863: 1282‐1297, 2016.
 38.Chan D, Lin J, Raffaniello RD. Expression and localization of rab escort protein isoforms in parotid acinar cells from rat. J Cell Physiol 185: 339‐347, 2000.
 39.Chen HQ, Tannous M, Veluthakal R, Amin R, Kowluru A. Novel roles for palmitoylation of Ras in IL‐1 beta‐induced nitric oxide release and caspase 3 activation in insulin‐secreting beta cells. Biochem Pharmacol 66: 1681‐1694, 2003.
 40.Cherfils J, Zeghouf M. Regulation of small GTPases by GEFs, GAPs, and GDIs. Physiol Rev 93: 269‐309, 2013.
 41.Cismowski MJ. Non‐receptor activators of heterotrimeric G protein signaling (AGS proteins). Semin Cell Dev Biol 17: 334‐344, 2006.
 42.Dai B, Zhang X, Shang R, Wang J, Yang X, Zhang H, Liu Q, Wang D, Wang L, Dou K. Blockade of ARHGAP11A reverses malignant progress via inactivating Rac1B in hepatocellular carcinoma. Cell Commun Signal 16: 99, 2018.
 43.Damacharla D, Thamilselvan V, Zhang X, Mestareehi A, Yi Z, Kowluru A. Quantitative proteomics reveals novel interaction partners of Rac1 in pancreatic beta‐cells: Evidence for increased interaction with Rac1 under hyperglycemic conditions. Mol Cell Endocrinol 494: 110489, 2019.
 44.Damoulakis G, Gambardella L, Rossman KL, Lawson CD, Anderson KE, Fukui Y, Welch HC, Der CJ, Stephens LR, Hawkins PT. P‐Rex1 directly activates RhoG to regulate GPCR‐driven Rac signalling and actin polarity in neutrophils. J Cell Sci 127: 2589‐2600, 2014.
 45.Day JW, Ottaway N, Patterson JT, Gelfanov V, Smiley D, Gidda J, Findeisen H, Bruemmer D, Drucker DJ, Chaudhary N, Holland J, Hembree J, Abplanalp W, Grant E, Ruehl J, Wilson H, Kirchner H, Lockie SH, Hofmann S, Woods SC, Nogueiras R, Pfluger PT, Perez‐Tilve D, DiMarchi R, Tschop MH. A new glucagon and GLP‐1 co‐agonist eliminates obesity in rodents. Nat Chem Biol 5: 749‐757, 2009.
 46.de Beco S, Vaidziulyte K, Manzi J, Dalier F, di Federico F, Cornilleau G, Dahan M, Coppey M. Optogenetic dissection of Rac1 and Cdc42 gradient shaping. Nat Commun 9: 4816, 2018.
 47.Deeney JT, Prentki M, Corkey BE. Metabolic control of beta‐cell function. Semin Cell Dev Biol 11: 267‐275, 2000.
 48.DerMardirossian C, Bokoch GM. GDIs: Central regulatory molecules in Rho GTPase activation. Trends Cell Biol 15: 356‐363, 2005.
 49.Dong H, Zhang Y, Wang J, Kim DS, Wu H, Sjogren B, Gao W, Luttrell L, Wang H. Regulator of G protein signaling 2 is a key regulator of pancreatic beta‐cell mass and function. Cell Death Dis 8: e2821, 2017.
 50.Dufurrena Q, Back N, Mains RE, Hodgson L, Tanowitz H, Mandela P, Eipper E, Kuliawat R. Kalirin/Trio Rho GDP/GTP exchange factors regulate proinsulin and insulin secretion. J Mol Endocrinol, 2018. DOI: 10.1530/JME‐18‐0048.
 51.Efrat S, Fleischer N, Hanahan D. Diabetes induced in male transgenic mice by expression of human H‐ras oncoprotein in pancreatic beta cells. Mol Cell Biol 10: 1779‐1783, 1990.
 52.El‐Sitt S, El‐Sibai M. The STAR of the DLC family. J Recept Signal Transduct Res 33: 10‐13, 2013.
 53.Evans JL, Goldfine ID, Maddux BA, Grodsky GM. Oxidative stress and stress‐activated signaling pathways: A unifying hypothesis of type 2 diabetes. Endocr Rev 23: 599‐622, 2002.
 54.Evcim AS, Micili SC, Karaman M, Erbil G, Guneli E, Gidener S, Gumustekin M. The role of Rac1 on carbachol‐induced contractile activity in detrusor smooth muscle from streptozotocin‐induced diabetic rats. Basic Clin Pharmacol Toxicol 116: 476‐484, 2015.
 55.Fahien LA, MacDonald MJ. The succinate mechanism of insulin release. Diabetes 51: 2669‐2676, 2002.
 56.Feng Q, Albeck JG, Cerione RA, Yang W. Regulation of the Cool/Pix proteins: Key binding partners of the Cdc42/Rac targets, the p21‐activated kinases. J Biol Chem 277: 5644‐5650, 2002.
 57.Fenske RJ, Cadena MT, Harenda QE, Wienkes HN, Carbajal K, Schaid MD, Laundre E, Brill AL, Truchan NA, Brar H, Wisinski J, Cai J, Graham TE, Engin F, Kimple ME. The inhibitory G protein α‐subunit, Gαz, promotes type 1 diabetes‐like pathophysiology in NOD mice. Endocrinology 158: 1645‐1658, 2017.
 58.Ferland RJ, Li X, Buhlmann JE, Bu X, Walsh CA, Lim B. Characterization of Rho‐GDIgamma and Rho‐GDIalpha mRNA in the developing and mature brain with an analysis of mice with targeted deletions of Rho‐GDIgamma. Brain Res 1054: 9‐21, 2005.
 59.Ferreira SM, Santos GJ, Rezende LF, Goncalves LM, Santos‐Silva JC, Bigarella CL, Carneiro EM, Saad ST, Boschero AC, Barbosa‐Sampaio HC. ARHGAP21 prevents abnormal insulin release through actin rearrangement in pancreatic islets from neonatal mice. Life Sci 127: 53‐58, 2015.
 60.Finan B, Yang B, Ottaway N, Smiley DL, Ma T, Clemmensen C, Chabenne J, Zhang L, Habegger KM, Fischer K, Campbell JE, Sandoval D, Seeley RJ, Bleicher K, Uhles S, Riboulet W, Funk J, Hertel C, Belli S, Sebokova E, Conde‐Knape K, Konkar A, Drucker DJ, Gelfanov V, Pfluger PT, Muller TD, Perez‐Tilve D, DiMarchi RD, Tschop MH. A rationally designed monomeric peptide triagonist corrects obesity and diabetes in rodents. Nat Med 21: 27‐36, 2015.
 61.Font de Mora J, Esteban LM, Burks DJ, Nunez A, Garces C, Garcia‐Barrado MJ, Iglesias‐Osma MC, Moratinos J, Ward JM, Santos E. Ras‐GRF1 signaling is required for normal beta‐cell development and glucose homeostasis. EMBO J 22: 3039‐3049, 2003.
 62.Fridlyand LE, Philipson LH. Oxidative reactive species in cell injury: Mechanisms in diabetes mellitus and therapeutic approaches. Ann N Y Acad Sci 1066: 136‐151, 2005.
 63.Fridlyand LE, Philipson LH. Pancreatic beta cell G protein coupled receptors and second messenger interactions: A systems biology computational analysis. PLoS One 11: e0152869, 2016.
 64.Gaisano HY. Here come the newcomer granules, better late than never. Trends Endocrinol Metab 25: 381‐388, 2014.
 65.Gao Y, Dickerson JB, Guo F, Zheng J, Zheng Y. Rational design and characterization of a Rac GTPase‐specific small molecule inhibitor. Proc Natl Acad Sci U S A 101: 7618‐7623, 2004.
 66.Garcia‐Mata R, Boulter E, Burridge K. The 'invisible hand': Regulation of RHO GTPases by RHOGDIs. Nat Rev Mol Cell Biol 12: 493‐504, 2011.
 67.Gault VA, Bhat VK, Irwin N, Flatt PR. A novel glucagon‐like peptide‐1 (GLP‐1)/glucagon hybrid peptide with triple‐acting agonist activity at glucose‐dependent insulinotropic polypeptide, GLP‐1, and glucagon receptors and therapeutic potential in high fat‐fed mice. J Biol Chem 288: 35581‐35591, 2013.
 68.Genier S, Degrandmaison J, Lavoie CL, Gendron L, Parent JL. Monitoring the aggregation of GPCRs by fluorescence microscopy. Methods Mol Biol 1947: 289‐302, 2019.
 69.Ghosh P, Rangamani P, Kufareva I. The GAPs, GEFs, GDIs and… now, GEMs: New kids on the heterotrimeric G protein signaling block. Cell Cycle 16: 607‐612, 2017.
 70.Gillingham AK, Bertram J, Begum F, Munro S. In vivo identification of GTPase interactors by mitochondrial relocalization and proximity biotinylation. Elife 8, 2019. DOI: 10.7554/eLife.45916.
 71.Gloerich M, Bos JL. Epac: Defining a new mechanism for cAMP action. Annu Rev Pharmacol Toxicol 50: 355‐375, 2010.
 72.Goalstone M, Kamath V, Kowluru A. Glucose activates prenyltransferases in pancreatic islet beta‐cells. Biochem Biophys Res Commun 391: 895‐898, 2010.
 73.Granata R, Settanni F, Trovato L, Gallo D, Gesmundo I, Nano R, Gallo MP, Bergandi L, Volante M, Alloatti G, Piemonti L, Leprince J, Papotti M, Vaudry H, Ong H, Ghigo E. RFamide peptides 43RFa and 26RFa both promote survival of pancreatic beta‐cells and human pancreatic islets but exert opposite effects on insulin secretion. Diabetes 63: 2380‐2393, 2014.
 74.Gross S, Devraj K, Feng Y, Macas J, Liebner S, Wieland T. Nucleoside diphosphate kinase B regulates angiogenic responses in the endothelium via caveolae formation and c‐Src‐mediated caveolin‐1 phosphorylation. J Cereb Blood Flow Metab 37: 2471‐2484, 2017.
 75.Groysman M, Hornstein I, Alcover A, Katzav S. Vav1 and Ly‐GDI two regulators of Rho GTPases, function cooperatively as signal transducers in T cell antigen receptor‐induced pathways. J Biol Chem 277: 50121‐50130, 2002.
 76.Groysman M, Russek CS, Katzav S. Vav, a GDP/GTP nucleotide exchange factor, interacts with GDIs, proteins that inhibit GDP/GTP dissociation. FEBS Lett 467: 75‐80, 2000.
 77.Gunton JE, Sisavanh M, Stokes RA, Satin J, Satin LS, Zhang M, Liu SM, Cai W, Cheng K, Cooney GJ, Laybutt DR, So T, Molero JC, Grey ST, Andres DA, Rolph MS, Mackay CR. Mice deficient in GEM GTPase show abnormal glucose homeostasis due to defects in beta‐cell calcium handling. PLoS One 7: e39462, 2012.
 78.Guo R, Jiang J, Jing Z, Chen Y, Shi Z, Deng B. Cysteinyl leukotriene receptor 1 regulates glucose‐stimulated insulin secretion (GSIS). Cell Signal 46: 129‐134, 2018.
 79.Hampton SE, Dore TM, Schmidt WK. Rce1: Mechanism and inhibition. Crit Rev Biochem Mol Biol 53: 157‐174, 2018.
 80.Hedman AC, Smith JM, Sacks DB. The biology of IQGAP proteins: Beyond the cytoskeleton. EMBO Rep 16: 427‐446, 2015.
 81.Hepler JR, Gilman AG. G proteins. Trends Biochem Sci 17: 383‐387, 1992.
 82.Higashijima T, Uzu S, Nakajima T, Ross EM. Mastoparan, a peptide toxin from wasp venom, mimics receptors by activating GTP‐binding regulatory proteins (G proteins). J Biol Chem 263: 6491‐6494, 1988.
 83.Hippe HJ, Wieland T. High energy phosphate transfer by NDPK B/Gbetagammacomplexes—An alternative signaling pathway involved in the regulation of basal cAMP production. J Bioenerg Biomembr 38: 197‐203, 2006.
 84.Hirose K, Kawashima T, Iwamoto I, Nosaka T, Kitamura T. MgcRacGAP is involved in cytokinesis through associating with mitotic spindle and midbody. J Biol Chem 276: 5821‐5828, 2001.
 85.Hodge RG, Ridley AJ. Regulating Rho GTPases and their regulators. Nat Rev Mol Cell Biol 17: 496‐510, 2016.
 86.Holz GG. Epac: A new cAMP‐binding protein in support of glucagon‐like peptide‐1 receptor‐mediated signal transduction in the pancreatic beta‐cell. Diabetes 53: 5‐13, 2004.
 87.Holz GG, Chepurny OG. Diabetes outfoxed by GLP‐1? Sci STKE 2005: pe2, 2005.
 88.Hoque M, Ali S, Hoda M. Current status of G protein coupled receptors as potential targets against type 2 diabetes mellitus. Int J Biol Macromol 118: 2237‐2244, 2018.
 89.Insel PA, Sriram K, Gorr MW, Wiley SZ, Michkov A, Salmeron C, Chinn AM. GPCRomics: An approach to discover GPCR drug targets. Trends Pharmacol Sci 40: 378‐387, 2019.
 90.Jayaram B, Syed I, Kyathanahalli CN, Rhodes CJ, Kowluru A. Arf nucleotide binding site opener [ARNO] promotes sequential activation of Arf6, Cdc42 and Rac1 and insulin secretion in INS 832/13 beta‐cells and rat islets. Biochem Pharmacol 81: 1016‐1027, 2011.
 91.Jayaram B, Syed I, Singh A, Subasinghe W, Kyathanahalli CN, Kowluru A. Isoprenylcysteine carboxyl methyltransferase facilitates glucose‐induced Rac1 activation, ROS generation and insulin secretion in INS 832/13 beta‐cells. Islets 3: 48‐57, 2011.
 92.Jensen MV, Gooding JR, Ferdaoussi M, Dai XQ, Peterson BS, PE MD, Newgard CB. Metabolomics applied to islet nutrient sensing mechanisms. Diabetes Obes Metab 19 (Suppl 1): 90‐94, 2017.
 93.Jezek P, Dlaskova A, Plecita‐Hlavata L. Redox homeostasis in pancreatic beta cells. Oxid Med Cell Longev 2012: 932838, 2012.
 94.Jiang H, Zhang X, Chen X, Aramsangtienchai P, Tong Z, Lin H. Protein lipidation: Occurrence, mechanisms, biological functions, and enabling technologies. Chem Rev 118: 919‐988, 2018.
 95.Jiang S, Shen D, Jia WJ, Han X, Shen N, Tao W, Gao X, Xue B, Li CJ. GGPPS‐mediated Rab27A geranylgeranylation regulates beta cell dysfunction during type 2 diabetes development by affecting insulin granule docked pool formation. J Pathol 238: 109‐119, 2016.
 96.Jitrapakdee S, Wutthisathapornchai A, Wallace JC, MacDonald MJ. Regulation of insulin secretion: Role of mitochondrial signalling. Diabetologia 53: 1019‐1032, 2010.
 97.Jonas KC, Hanyaloglu AC. Analysis of spatial assembly of GPCRs using photoactivatable dyes and localization microscopy. Methods Mol Biol 1947: 337‐348, 2019.
 98.Jong YI, Harmon SK, O'Malley KL. Intracellular GPCRs play key roles in synaptic plasticity. ACS Chem Nerosci 9: 2162‐2172, 2018.
 99.Kagawa Y, Matsumoto S, Kamioka Y, Mimori K, Naito Y, Ishii T, Okuzaki D, Nishida N, Maeda S, Naito A, Kikuta J, Nishikawa K, Nishimura J, Haraguchi N, Takemasa I, Mizushima T, Ikeda M, Yamamoto H, Sekimoto M, Ishii H, Doki Y, Matsuda M, Kikuchi A, Mori M, Ishii M. Cell cycle‐dependent Rho GTPase activity dynamically regulates cancer cell motility and invasion in vivo. PLoS One 8: e83629, 2013.
 100.Kalwat MA, Wiseman DA, Luo W, Wang Z, Thurmond DC. Gelsolin associates with the N terminus of syntaxin 4 to regulate insulin granule exocytosis. Mol Endocrinol 26: 128‐141, 2012.
 101.Karunakaran U, Park KG. A systematic review of oxidative stress and safety of antioxidants in diabetes: Focus on islets and their defense. Diabetes Metab J 37: 106‐112, 2013.
 102.Kasai K, Ohara‐Imaizumi M, Takahashi N, Mizutani S, Zhao S, Kikuta T, Kasai H, Nagamatsu S, Gomi H, Izumi T. Rab27a mediates the tight docking of insulin granules onto the plasma membrane during glucose stimulation. J Clin Invest 115: 388‐396, 2005.
 103.Kashima Y, Miki T, Shibasaki T, Ozaki N, Miyazaki M, Yano H, Seino S. Critical role of cAMP‐GEFII‐Rim2 complex in incretin‐potentiated insulin secretion. J Biol Chem 276: 46046‐46053, 2001.
 104.Kawashima T, Bao YC, Nomura Y, Moon Y, Tonozuka Y, Minoshima Y, Hatori T, Tsuchiya A, Kiyono M, Nosaka T, Nakajima H, Williams DA, Kitamura T. Rac1 and a GTPase‐activating protein, MgcRacGAP, are required for nuclear translocation of STAT transcription factors. J Cell Biol 175: 937‐946, 2006.
 105.Keane KN, Cruzat VF, Carlessi R, de Bittencourt PI Jr, Newsholme P. Molecular events linking oxidative stress and inflammation to insulin resistance and beta‐cell dysfunction. Oxid Med Cell Longev 181643: 2015, 2015.
 106.Kebede MA, Alquier T, Latour MG, Poitout V. Lipid receptors and islet function: Therapeutic implications? Diabetes Obes Metab 11 (Suppl 4): 10‐20, 2009.
 107.Kepner EM, Yoder SM, Oh E, Kalwat MA, Wang Z, Quilliam LA, Thurmond DC. Cool‐1/betaPIX functions as a guanine nucleotide exchange factor in the cycling of Cdc42 to regulate insulin secretion. Am J Physiol Endocrinol Metab 301: E1072‐E1080, 2011.
 108.Khajavi N, Finan B, Kluth O, Muller TD, Mergler S, Schulz A, Kleinau G, Scheerer P, Schurmann A, Gudermann T, Tschop MH, Krude H, DiMarchi RD, Biebermann H. An incretin‐based tri‐agonist promotes superior insulin secretion from murine pancreatic islets via PLC activation. Cell Signal 51: 13‐22, 2018.
 109.Khan SM, Sleno R, Gora S, Zylbergold P, Laverdure JP, Labbe JC, Miller GJ, Hebert TE. The expanding roles of Gbetagamma subunits in G protein‐coupled receptor signaling and drug action. Pharmacol Rev 65: 545‐577, 2013.
 110.Khan SM, Sung JY, Hebert TE. Gbetagamma subunits—Different spaces, different faces. Pharmacol Res 111: 434‐441, 2016.
 111.Khoo S, Gibson TB, Arnette D, Lawrence M, January B, McGlynn K, Vanderbilt CA, Griffen SC, German MS, Cobb MH. MAP kinases and their roles in pancreatic beta‐cells. Cell Biochem Biophys 40: 191‐200, 2004.
 112.Khoo S, Griffen SC, Xia Y, Baer RJ, German MS, Cobb MH. Regulation of insulin gene transcription by ERK1 and ERK2 in pancreatic beta cells. J Biol Chem 278: 32969‐32977, 2003.
 113.Kobilka B. The structural basis of G protein‐coupled receptor signaling (Nobel Lecture). Angew Chem Int Ed Engl 52: 6380‐6388, 2013.
 114.Koelle MR. A new family of G protein regulators – The RGS proteins. Curr Opin Cell Biol 9: 143‐147, 1997.
 115.Komatsu M, McDermott AM, Gillison SL, Sharp GW. Mastoparan stimulates exocytosis at a Ca(2+)‐independent late site in stimulus‐secretion coupling. Studies with the RINm5F beta‐cell line. J Biol Chem 268: 23297‐23306, 1993.
 116.Komatsu M, Takei M, Ishii H, Sato Y. Glucose‐stimulated insulin secretion: A newer perspective. J Diabetes Investig 4: 511‐516, 2013.
 117.Kong M, Bui TV, Ditsworth D, Gruber JJ, Goncharov D, Krymskaya VP, Lindsten T, Thompson CB. The PP2A‐associated protein alpha4 plays a critical role in the regulation of cell spreading and migration. J Biol Chem 282: 29712‐29720, 2007.
 118.Kong X, Yan D, Sun J, Wu X, Mulder H, Hua X, Ma X. Glucagon‐like peptide 1 stimulates insulin secretion via inhibiting RhoA/ROCK signaling and disassembling glucotoxicity‐induced stress fibers. Endocrinology 155: 4676‐4685, 2014.
 119.Konrad RJ, Young RA, Record RD, Smith RM, Butkerait P, Manning D, Jarett L, Wolf BA. The heterotrimeric G protein Gi is localized to the insulin secretory granules of beta‐cells and is involved in insulin exocytosis. J Biol Chem 270: 12869‐12876, 1995.
 120.Koo TH, Eipper BA, Donaldson JG. Arf6 recruits the Rac GEF Kalirin to the plasma membrane facilitating Rac activation. BMC Cell Biol 8: 29, 2007.
 121.Kowluru A. Adenine and guanine nucleotide‐specific succinyl‐CoA synthetases in the clonal beta‐cell mitochondria: Implications in the beta‐cell high‐energy phosphate metabolism in relation to physiological insulin secretion. Diabetologia 44: 89‐94, 2001.
 122.Kowluru A. Identification and characterization of a novel protein histidine kinase in the islet beta cell: Evidence for its regulation by mastoparan, an activator of G proteins and insulin secretion. Biochem Pharmacol 63: 2091‐2100, 2002.
 123.Kowluru A. Defective protein histidine phosphorylation in islets from the Goto‐Kakizaki diabetic rat. Am J Physiol Endocrinol Metab 285: E498‐E503, 2003.
 124.Kowluru A. Regulatory roles for small G proteins in the pancreatic beta‐cell: Lessons from models of impaired insulin secretion. Am J Physiol Endocrinol Metab 285: E669‐E684, 2003.
 125.Kowluru A. Emerging roles for protein histidine phosphorylation in cellular signal transduction: Lessons from the islet beta‐cell. J Cell Mol Med 12: 1885‐1908, 2008.
 126.Kowluru A. Protein prenylation in glucose‐induced insulin secretion from the pancreatic islet beta cell: A perspective. J Cell Mol Med 12: 164‐173, 2008.
 127.Kowluru A. Small G proteins in islet beta‐cell function. Endocr Rev 31: 52‐78, 2010.
 128.Kowluru A. Friendly, and not so friendly, roles of Rac1 in islet beta‐cell function: Lessons learnt from pharmacological and molecular biological approaches. Biochem Pharmacol 81: 965‐975, 2011.
 129.Kowluru A. A lack of ‘glue’ misplaces Rab27A to cause islet dysfunction in diabetes. J Pathol 238: 375‐377, 2016.
 130.Kowluru A. Inappropriate movement of Rac1 contributes to glucotoxicity of the islet beta‐cell. Cell Cycle 16: 1387‐1388, 2017.
 131.Kowluru A. Role of G proteins in islet function in health and diabetes. Diabetes Obes Metab 19 (Suppl 1): 63‐75, 2017.
 132.Kowluru A. Tiam1/Vav2‐Rac1 axis: A tug‐of‐war between islet function and dysfunction. Biochem Pharmacol 132: 9‐17, 2017.
 133.Kowluru A. Novel roles of alpha‐4, non‐canonical scaffolding subunit of protein phosphatase 2A, in the onset of beta cell dysfunction under glucotoxic conditions. Diabetologia 61: S1‐S620, 2018.
 134.Kowluru A, Amin R. Inhibitors of post‐translational modifications of G proteins as probes to study the pancreatic beta cell function: Potential therapeutic implications. Curr Drug Targets Immune Endocr Metabol Disord 2: 129‐139, 2002.
 135.Kowluru A, Klumpp S, Krieglstein J. Protein histidine [de]phosphorylation in insulin secretion: Abnormalities in models of impaired insulin secretion. Naunyn Schmiedebergs Arch Pharmacol 384: 383‐390, 2011.
 136.Kowluru A, Kowluru RA. Phagocyte‐like NADPH oxidase [Nox2] in cellular dysfunction in models of glucolipotoxicity and diabetes. Biochem Pharmacol 88: 275‐283, 2014.
 137.Kowluru A, Kowluru RA. Protein prenylation in islet beta‐cell function in health and diabetes: Putting the pieces of the puzzle together. Biochem Pharmacol 98: 363‐370, 2015.
 138.Kowluru A, Kowluru RA. RACking up ceramide‐induced islet beta‐cell dysfunction. Biochem Pharmacol 154: 161‐169, 2018.
 139.Kowluru A, Li G, Metz SA. Glucose activates the carboxyl methylation of gamma subunits of trimeric GTP‐binding proteins in pancreatic beta cells. Modulation in vivo by calcium, GTP, and pertussis toxin. J Clin Invest 100: 1596‐1610, 1997.
 140.Kowluru A, Li G, Rabaglia ME, Segu VB, Hofmann F, Aktories K, Metz SA. Evidence for differential roles of the Rho subfamily of GTP‐binding proteins in glucose‐ and calcium‐induced insulin secretion from pancreatic beta cells. Biochem Pharmacol 54: 1097‐1108, 1997.
 141.Kowluru A, Metz SA. Characterization of nucleoside diphosphokinase activity in human and rodent pancreatic beta cells: Evidence for its role in the formation of guanosine triphosphate, a permissive factor for nutrient‐induced insulin secretion. Biochemistry 33: 12495‐12503, 1994.
 142.Kowluru A, Metz SA. Regulation of guanine‐nucleotide binding proteins in islet subcellular fractions by phospholipase‐derived lipid mediators of insulin secretion. Biochim Biophys Acta 1222: 360‐368, 1994.
 143.Kowluru A, Metz SA. Stimulation by prostaglandin E2 of a high‐affinity GTPase in the secretory granules of normal rat and human pancreatic islets. Biochem J 297 (Pt 2): 399‐406, 1994.
 144.Kowluru A, Seavey SE, Li G, Sorenson RL, Weinhaus AJ, Nesher R, Rabaglia ME, Vadakekalam J, Metz SA. Glucose‐ and GTP‐dependent stimulation of the carboxyl methylation of CDC42 in rodent and human pancreatic islets and pure beta cells. Evidence for an essential role of GTP‐binding proteins in nutrient‐induced insulin secretion. J Clin Invest 98: 540‐555, 1996.
 145.Kowluru A, Seavey SE, Rhodes CJ, Metz SA. A novel regulatory mechanism for trimeric GTP‐binding proteins in the membrane and secretory granule fractions of human and rodent beta cells. Biochem J 313 (Pt 1): 97‐107, 1996.
 146.Kowluru A, Tannous M, Chen HQ. Localization and characterization of the mitochondrial isoform of the nucleoside diphosphate kinase in the pancreatic beta cell: Evidence for its complexation with mitochondrial succinyl‐CoA synthetase. Arch Biochem Biophys 398: 160‐169, 2002.
 147.Kowluru A, Veluthakal R. Rho guanosine diphosphate‐dissociation inhibitor plays a negative modulatory role in glucose‐stimulated insulin secretion. Diabetes 54: 3523‐3529, 2005.
 148.Kowluru A, Veluthakal R, Kaetzel DM. Regulatory roles for nm23/nucleoside diphosphate kinase‐like enzymes in insulin secretion from the pancreatic islet beta cell. J Bioenerg Biomembr 38: 227‐232, 2006.
 149.Kowluru A, Veluthakal R, Rhodes CJ, Kamath V, Syed I, Koch BJ. Protein farnesylation‐dependent Raf/extracellular signal‐related kinase signaling links to cytoskeletal remodeling to facilitate glucose‐induced insulin secretion in pancreatic beta‐cells. Diabetes 59: 967‐977, 2010.
 150.Kowluru RA, Kowluru A, Veluthakal R, Mohammad G, Syed I, Santos JM, Mishra M. TIAM1‐RAC1 signalling axis‐mediated activation of NADPH oxidase‐2 initiates mitochondrial damage in the development of diabetic retinopathy. Diabetologia 57: 1047‐1056, 2014.
 151.Langmead CJ, Summers RJ. Molecular pharmacology of GPCRs. Br J Pharmacol 175: 4005‐4008, 2018.
 152.Lawrence JT, Birnbaum MJ. ADP‐ribosylation factor 6 regulates insulin secretion through plasma membrane phosphatidylinositol 4,5‐bisphosphate. Proc Natl Acad Sci U S A 100: 13320‐13325, 2003.
 153.Lawrence MC, McGlynn K, Park BH, Cobb MH. ERK1/2‐dependent activation of transcription factors required for acute and chronic effects of glucose on the insulin gene promoter. J Biol Chem 280: 26751‐26759, 2005.
 154.Lawson CD, Der CJ. Filling GAPs in our knowledge: ARHGAP11A and RACGAP1 act as oncogenes in basal‐like breast cancers. Small GTPases 9: 290‐296, 2018.
 155.Lawson CD, Fan C, Mitin N, Baker NM, George SD, Graham DM, Perou CM, Burridge K, Der CJ, Rossman KL. Rho GTPase transcriptome analysis reveals oncogenic roles for Rho GTPase‐activating proteins in basal‐like breast cancers. Cancer Res 76: 3826‐3837, 2016.
 156.Lee JJ, Kim HS, Lee JS, Park J, Shin SC, Song S, Lee E, Choi JE, Suh JW, Lee H, Kim EE, Seo EK, Shin DH, Lee HY, Lee KJ. Small molecule activator of Nm23/NDPK as an inhibitor of metastasis. Sci Rep 8: 10909, 2018.
 157.Lee YS, Jun HS. Anti‐diabetic actions of glucagon‐like peptide‐1 on pancreatic beta‐cells. Metabolism 63: 9‐19, 2014.
 158.Leech CA, Holz GG, Chepurny O, Habener JF. Expression of cAMP‐regulated guanine nucleotide exchange factors in pancreatic beta‐cells. Biochem Biophys Res Commun 278: 44‐47, 2000.
 159.Lefkowitz RJ. A brief history of G protein coupled receptors (Nobel Lecture). Angew Chem Int Ed Engl 52: 6366‐6378, 2013.
 160.Leiser M, Efrat S, Fleischer N. Evidence that Rap1 carboxylmethylation is involved in regulated insulin secretion. Endocrinology 136: 2521‐2530, 1995.
 161.Lenzen S. Chemistry and biology of reactive species with special reference to the antioxidative defence status in pancreatic beta‐cells. Biochim Biophys Acta Gen Subj 1861: 1929‐1942, 2017.
 162.Li G, Regazzi R, Roche E, Wollheim CB. Blockade of mevalonate production by lovastatin attenuates bombesin and vasopressin potentiation of nutrient‐induced insulin secretion in HIT‐T15 cells. Probable involvement of small GTP‐binding proteins. Biochem J 289 (Pt 2): 379‐385, 1993.
 163.Li H, Liu T, Lim J, Gounko NV, Hong W, Han W. Increased biogenesis of glucagon‐containing secretory granules and glucagon secretion in BIG3‐knockout mice. Mol Metab 4: 246‐252, 2015.
 164.Li H, Wei S, Cheng K, Gounko NV, Ericksen RE, Xu A, Hong W, Han W. BIG3 inhibits insulin granule biogenesis and insulin secretion. EMBO Rep 15: 714‐722, 2014.
 165.Liao J, Ye Z, Huang G, Xu C, Guo Q, Wang E. Delayed treatment with NSC23766 in streptozotocin‐induced diabetic rats ameliorates post‐ischemic neuronal apoptosis through suppression of mitochondrial p53 translocation. Neuropharmacology 85: 508‐516, 2014.
 166.Ljubicic S, Bezzi P, Brajkovic S, Nesca V, Guay C, Ohbayashi N, Fukuda M, Abderrhamani A, Regazzi R. The GTPase Rab37 participates in the control of insulin exocytosis. PLoS One 8: e68255, 2013.
 167.Lu W, Wang J, Wen T. Downregulation of Rho‐GDI gamma promotes differentiation of neural stem cells. Mol Cell Biochem 311: 233‐240, 2008.
 168.Lundius EG, Vukojevic V, Hertz E, Stroth N, Cederlund A, Hiraiwa M, Terenius L, Svenningsson P. GPR37 protein trafficking to the plasma membrane regulated by prosaposin and GM1 gangliosides promotes cell viability. J Biol Chem 289: 4660‐4673, 2014.
 169.MacDonald MJ. Elusive proximal signals of beta‐cells for insulin secretion. Diabetes 39: 1461‐1466, 1990.
 170.Maechler P. Mitochondrial function and insulin secretion. Mol Cell Endocrinol 379: 12‐18, 2013.
 171.Magnani F, Mattevi A. Structure and mechanisms of ROS generation by NADPH oxidases. Curr Opin Struct Biol 59: 91‐97, 2019.
 172.Malaisse WJ. Insulin release: The receptor hypothesis. Diabetologia 57: 1287‐1290, 2014.
 173.Manser E, Loo TH, Koh CG, Zhao ZS, Chen XQ, Tan L, Tan I, Leung T, Lim L. PAK kinases are directly coupled to the PIX family of nucleotide exchange factors. Mol Cell 1: 183‐192, 1998.
 174.Marei H, Carpy A, Macek B, Malliri A. Proteomic analysis of Rac1 signaling regulation by guanine nucleotide exchange factors. Cell Cycle 15: 1961‐1974, 2016.
 175.Marei H, Malliri A. GEFs: Dual regulation of Rac1 signaling. Small GTPases 8: 90‐99, 2017.
 176.Marei H, Malliri A. Rac1 in human diseases: The therapeutic potential of targeting Rac1 signaling regulatory mechanisms. Small GTPases 8: 139‐163, 2017.
 177.McDonald P, Veluthakal R, Kaur H, Kowluru A. Biologically active lipids promote trafficking and membrane association of Rac1 in insulin‐secreting INS 832/13 cells. Am J Physiol Cell Physiol 292: C1216‐C1220, 2007.
 178.Metz SA. Membrane phospholipid turnover as an intermediary step in insulin secretion. Putative roles of phospholipases in cell signaling. Am J Med 85: 9‐21, 1988.
 179.Metz SA. The pancreatic islet as Rubik's Cube. Is phospholipid hydrolysis a piece of the puzzle? Diabetes 40: 1565‐1573, 1991.
 180.Metz SA, Kowluru A. Inosine monophosphate dehydrogenase: A molecular switch integrating pleiotropic GTP‐dependent beta‐cell functions. Proc Assoc Am Physicians 111: 335‐346, 1999.
 181.Metz SA, Meredith M, Rabaglia ME, Kowluru A. Small elevations of glucose concentration redirect and amplify the synthesis of guanosine 5′‐triphosphate in rat islets. J Clin Invest 92: 872‐882, 1993.
 182.Metz SA, Meredith M, Vadakekalam J, Rabaglia ME, Kowluru A. A defect late in stimulus‐secretion coupling impairs insulin secretion in Goto‐Kakizaki diabetic rats. Diabetes 48: 1754‐1762, 1999.
 183.Metz SA, Rabaglia ME, Pintar TJ. Selective inhibitors of GTP synthesis impede exocytotic insulin release from intact rat islets. J Biol Chem 267: 12517‐12527, 1992.
 184.Metz SA, Rabaglia ME, Stock JB, Kowluru A. Modulation of insulin secretion from normal rat islets by inhibitors of the post‐translational modifications of GTP‐binding proteins. Biochem J 295 (Pt 1): 31‐40, 1993.
 185.Milligan G, Kostenis E. Heterotrimeric G proteins: A short history. Br J Pharmacol 147 (Suppl 1): S46‐S55, 2006.
 186.Mizrahi A, Molshanski‐Mor S, Weinbaum C, Zheng Y, Hirshberg M, Pick E. Activation of the phagocyte NADPH oxidase by Rac Guanine nucleotide exchange factors in conjunction with ATP and nucleoside diphosphate kinase. J Biol Chem 280: 3802‐3811, 2005.
 187.Mohammad G, Duraisamy AJ, Kowluru A, Kowluru RA. Functional regulation of an oxidative stress mediator, Rac1, in diabetic retinopathy. Mol Neurobiol 56 (12): 8643‐8655, 2019.
 188.Mohammed AM, Syeda K, Hadden T, Kowluru A. Upregulation of phagocyte‐like NADPH oxidase by cytokines in pancreatic beta‐cells: Attenuation of oxidative and nitrosative stress by 2‐bromopalmitate. Biochem Pharmacol 85: 109‐114, 2013.
 189.Morgan D, Rebelato E, Abdulkader F, Graciano MF, Oliveira‐Emilio HR, Hirata AE, Rocha MS, Bordin S, Curi R, Carpinelli AR. Association of NAD(P)H oxidase with glucose‐induced insulin secretion by pancreatic beta‐cells. Endocrinology 150: 2197‐2201, 2009.
 190.Mukai E, Fujimoto S, Sato H, Oneyama C, Kominato R, Sato Y, Sasaki M, Nishi Y, Okada M, Inagaki N. Exendin‐4 suppresses SRC activation and reactive oxygen species production in diabetic Goto‐Kakizaki rat islets in an Epac‐dependent manner. Diabetes 60: 218‐226, 2011.
 191.Murakami T, Shoji M, Imai Y, Inoue H, Kawarabayashi T, Matsubara E, Harigaya Y, Sasaki A, Takahashi R, Abe K. Pael‐R is accumulated in Lewy bodies of Parkinson's disease. Ann Neurol 55: 439‐442, 2004.
 192.Nauert JB, Rigas JD, Lester LB. Identification of an IQGAP1/AKAP79 complex in beta‐cells. J Cell Biochem 90: 97‐108, 2003.
 193.Naumann H, Rathjen T, Poy MN, Spagnoli FM. The RhoGAP Stard13 controls insulin secretion through F‐actin remodeling. Mol Metab 8: 96‐105, 2018.
 194.Neer EJ. Heterotrimeric G proteins: Organizers of transmembrane signals. Cell 80: 249‐257, 1995.
 195.Nelson TY, Boyd AE III. Gelsolin, a Ca2+‐dependent actin‐binding protein in a hamster insulin‐secreting cell line. J Clin Invest 75: 1015‐1022, 1985.
 196.Nevins AK, Thurmond DC. Glucose regulates the cortical actin network through modulation of Cdc42 cycling to stimulate insulin secretion. Am J Physiol Cell Physiol 285: C698‐C710, 2003.
 197.Nevins AK, Thurmond DC. Caveolin‐1 functions as a novel Cdc42 guanine nucleotide dissociation inhibitor in pancreatic beta‐cells. J Biol Chem 281: 18961‐18972, 2006.
 198.Newgard CB, McGarry JD. Metabolic coupling factors in pancreatic beta‐cell signal transduction. Annu Rev Biochem 64: 689‐719, 1995.
 199.Newsholme P, Keane KN, Carlessi R, Cruzat V. Oxidative stress pathways in pancreatic beta cells and insulin sensitive cells and tissues – Importance to cell metabolism, function and dysfunction. Am J Physiol Cell Physiol 317 (3): C420‐C433, 2019.
 200.Newsholme P, Morgan D, Rebelato E, Oliveira‐Emilio HC, Procopio J, Curi R, Carpinelli A. Insights into the critical role of NADPH oxidase(s) in the normal and dysregulated pancreatic beta cell. Diabetologia 52: 2489‐2498, 2009.
 201.Nicholls DG. The pancreatic beta‐cell: A bioenergetic perspective. Physiol Rev 96: 1385‐1447, 2016.
 202.Nouri K, Timson DJ, Ahmadian MR. New model for the interaction of IQGAP1 with CDC42 and RAC1. Small GTPases: 1‐7, 2017. DOI: 10.1080/21541248.2017.1321169.
 203.Oh DY, Olefsky JM. G protein‐coupled receptors as targets for anti‐diabetic therapeutics. Nat Rev Drug Discov 15: 161‐172, 2016.
 204.Oh YS, Lee YJ, Kang Y, Han J, Lim OK, Jun HS. Exendin‐4 inhibits glucolipotoxic ER stress in pancreatic beta cells via regulation of SREBP1c and C/EBPbeta transcription factors. J Endocrinol 216: 343‐352, 2013.
 205.Olaniru OE, Persaud SJ. Adhesion G protein coupled receptors: Implications for metabolic function. Pharmacol Ther 198: 123‐134, 2019.
 206.Olate J, Allende JE. Structure and function of G proteins. Pharmacol Ther 51: 403‐419, 1991.
 207.Olofsson B. Rho guanine dissociation inhibitors: Pivotal molecules in cellular signalling. Cell Signal 11: 545‐554, 1999.
 208.Orlov DN, Orlov N. Nucleoside diphosphate kinase and GTP‐binding proteins. Possible mechanisms of coupling. Biofizika 53: 922‐928, 2008.
 209.Paglialunga S, Simnett G, Robson H, Hoang M, Pillai R, Arkell AM, Simpson JA, Bonen A, Huising M, Joseph JW, Holloway GP. The Rab‐GTPase activating protein, TBC1D1, is critical for maintaining normal glucose homeostasis and beta‐cell mass. Appl Physiol Nutr Metab 42: 647‐655, 2017.
 210.Pantarelli C, Welch HCE. Rac‐GTPases and Rac‐GEFs in neutrophil adhesion, migration and recruitment. Eur J Clin Invest 48 (Suppl 2): e12939, 2018.
 211.Park F. Accessory proteins for heterotrimeric G proteins in the kidney. Front Physiol 6: 219, 2015.
 212.Park F. Activators of G protein signaling in the kidney. J Pharmacol Exp Ther 353: 235‐245, 2015.
 213.Parton RG, del Pozo MA. Caveolae as plasma membrane sensors, protectors and organizers. Nat Rev Mol Cell Biol 14: 98‐112, 2013.
 214.Persaud SJ. Islet G protein coupled receptors: Therapeutic potential for diabetes. Curr Opin Pharmacol 37: 24‐28, 2017.
 215.Perschbacher KJ, Deng G, Fisher RA, Gibson‐Corley KN, Santillan MK, Grobe JL. Regulators of G protein signaling in cardiovascular function during pregnancy. Physiol Genomics 50: 590‐604, 2018.
 216.Plecita‐Hlavata L, Engstova H, Jezek J, Holendova B, Tauber J, Petraskova L, Kren V, Jezek P. Potential of mitochondria‐targeted antioxidants to prevent oxidative stress in pancreatic beta‐cells. Oxid Med Cell Longev 2019: 1826303, 2019.
 217.Prentki M, Matschinsky FM, Madiraju SR. Metabolic signaling in fuel‐induced insulin secretion. Cell Metab 18: 162‐185, 2013.
 218.Quitterer U, AbdAlla S. Discovery of pathologic GPCR aggregation. Front Med (Lausanne) 6: 9, 2019.
 219.Rathor N, Chung HK, Wang SR, Qian M, Turner DJ, Wang JY, Rao JN. beta‐PIX plays an important role in regulation of intestinal epithelial restitution by interacting with GIT1 and Rac1 after wounding. Am J Physiol Gastrointest Liver Physiol 314: G399‐G407, 2018.
 220.Regazzi R, Kikuchi A, Takai Y, Wollheim CB. The small GTP‐binding proteins in the cytosol of insulin‐secreting cells are complexed to GDP dissociation inhibitor proteins. J Biol Chem 267: 17512‐17519, 1992.
 221.Reimann F, Gribble FM. G protein‐coupled receptors as new therapeutic targets for type 2 diabetes. Diabetologia 59: 229‐233, 2016.
 222.Rives ML, Rady B, Swanson N, Zhao S, Qi J, Arnoult E, Bakaj I, Mancini A, Breton B, Lee SP, Player MR, Pocai A. GPR40‐mediated Galpha12 activation by allosteric full agonists highly efficacious at potentiating glucose‐stimulated insulin secretion in human islets. Mol Pharmacol 93: 581‐591, 2018.
 223.Robertson RP. Antioxidant drugs for treating beta‐cell oxidative stress in type 2 diabetes: Glucose‐centric versus insulin‐centric therapy. Discov Med 9: 132‐137, 2010.
 224.Robertson RP, Harmon J, Tran PO, Poitout V. Beta‐cell glucose toxicity, lipotoxicity, and chronic oxidative stress in type 2 diabetes. Diabetes 53 (Suppl 1): S119‐S124, 2004.
 225.Robertson RP, Seaquist ER, Walseth TF. G proteins and modulation of insulin secretion. Diabetes 40: 1‐6, 1991.
 226.Rodriguez‐Fdez S, Bustelo XR. The Vav GEF family: An evolutionary and functional perspective. Cells 8, 2019. DOI: 10.3390/cells8050465.
 227.Rorsman P, Ramracheya R, Rorsman NJ, Zhang Q. ATP‐regulated potassium channels and voltage‐gated calcium channels in pancreatic alpha and beta cells: Similar functions but reciprocal effects on secretion. Diabetologia 57: 1749‐1761, 2014.
 228.Rosa LRO, Soares GM, Silveira LR, Boschero AC, Barbosa‐Sampaio HCL. ARHGAP21 as a master regulator of multiple cellular processes. J Cell Physiol 233: 8477‐8481, 2018.
 229.Rosenbaum DM, Rasmussen SG, Kobilka BK. The structure and function of G protein‐coupled receptors. Nature 459: 356‐363, 2009.
 230.Rothe J, Thor D, Winkler J, Knierim AB, Binder C, Huth S, Kraft R, Rothemund S, Schoneberg T, Promel S. Involvement of the adhesion GPCRs latrophilins in the regulation of insulin release. Cell Rep 26: 1573‐1584, 2019.
 231.Ruiz de Azua I, Scarselli M, Rosemond E, Gautam D, Jou W, Gavrilova O, Ebert PJ, Levitt P, Wess J. RGS4 is a negative regulator of insulin release from pancreatic beta‐cells in vitro and in vivo. Proc Natl Acad Sci U S A 107: 7999‐8004, 2010.
 232.Sahajpal N, Kowluru A, Kowluru RA. The regulatory role of Rac1, a small molecular weight GTPase, in the development of diabetic retinopathy. J Clin Med 8, 2019. DOI: 10.3390/jcm8070965.
 233.Sandrock K, Bielek H, Schradi K, Schmidt G, Klugbauer N. The nuclear import of the small GTPase Rac1 is mediated by the direct interaction with karyopherin alpha2. Traffic 11: 198‐209, 2010.
 234.Sato Y, Fujimoto S, Mukai E, Sato H, Tahara Y, Ogura K, Yamano G, Ogura M, Nagashima K, Inagaki N. Palmitate induces reactive oxygen species production and beta‐cell dysfunction by activating nicotinamide adenine dinucleotide phosphate oxidase through Src signaling. J Diabetes Investig 5: 19‐26, 2014.
 235.Schiattarella GG, Carrizzo A, Ilardi F, Damato A, Ambrosio M, Madonna M, Trimarco V, Marino M, De Angelis E, Settembrini S, Perrino C, Trimarco B, Esposito G, Vecchione C. Rac1 modulates endothelial function and platelet aggregation in diabetes mellitus. J Am Heart Assoc 7, 2018. DOI: 10.1161/JAHA.117.007322.
 236.Schmidt S, Debant A. Function and regulation of the Rho guanine nucleotide exchange factor Trio. Small GTPases 5: e29769, 2014.
 237.Seaquist ER, Walseth TF, Redmon JB, Robertson RP. G protein regulation of insulin secretion. J Lab Clin Med 123: 338‐345, 1994.
 238.Seino S, Takahashi H, Fujimoto W, Shibasaki T. Roles of cAMP signalling in insulin granule exocytosis. Diabetes Obes Metab 11 (Suppl 4): 180‐188, 2009.
 239.Senese NB, Rasenick MM, Traynor JR. The role of G proteins and G protein regulating proteins in depressive disorders. Front Pharmacol 9: 1289, 2018.
 240.Seno K, Hayashi F. Palmitoylation is a prerequisite for dimerization‐dependent raftophilicity of rhodopsin. J Biol Chem 292: 15321‐15328, 2017.
 241.Serafimidis I, Heximer S, Beis D, Gavalas A. G protein‐coupled receptor signaling and sphingosine‐1‐phosphate play a phylogenetically conserved role in endocrine pancreas morphogenesis. Mol Cell Biol 31: 4442‐4453, 2011.
 242.Sethakorn N, Yau DM, Dulin NO. Non‐canonical functions of RGS proteins. Cell Signal 22: 1274‐1281, 2010.
 243.Shan L, Li J, Wei M, Ma J, Wan L, Zhu W, Li Y, Zhu H, Arnold JM, Peng T. Disruption of Rac1 signaling reduces ischemia‐reperfusion injury in the diabetic heart by inhibiting calpain. Free Radic Biol Med 49: 1804‐1814, 2010.
 244.Sharp GW. Mechanisms of inhibition of insulin release. Am J Physiol 271: C1781‐C1799, 1996.
 245.Shen E, Li Y, Li Y, Shan L, Zhu H, Feng Q, Arnold JM, Peng T. Rac1 is required for cardiomyocyte apoptosis during hyperglycemia. Diabetes 58: 2386‐2395, 2009.
 246.Shibasaki T, Takahashi T, Takahashi H, Seino S. Cooperation between cAMP signalling and sulfonylurea in insulin secretion. Diabetes Obes Metab 16 (Suppl 1): 118‐125, 2014.
 247.Shou C, Farnsworth CL, Neel BG, Feig LA. Molecular cloning of cDNAs encoding a guanine‐nucleotide‐releasing factor for Ras p21. Nature 358: 351‐354, 1992.
 248.Shou C, Wurmser A, Suen KL, Barbacid M, Feig LA, Ling K. Differential response of the Ras exchange factor, Ras‐GRF to tyrosine kinase and G protein mediated signals. Oncogene 10: 1887‐1893, 1995.
 249.Sidarala V, Kowluru A. Exposure to chronic hyperglycemic conditions results in Ras‐related C3 botulinum toxin substrate 1 (Rac1)‐mediated activation of p53 and ATM kinase in pancreatic beta‐cells. Apoptosis 22: 597‐607, 2017.
 250.Sidarala V, Kowluru A. The regulatory roles of mitogen‐activated protein kinase (MAPK) pathways in health and diabetes: Lessons learned from the pancreatic beta‐cell. Recent Pat Endocr Metab Immune Drug Discov 10: 76‐84, 2017.
 251.Sidarala V, Veluthakal R, Syeda K, Vlaar C, Newsholme P, Kowluru A. Phagocyte‐like NADPH oxidase (Nox2) promotes activation of p38MAPK in pancreatic beta‐cells under glucotoxic conditions: Evidence for a requisite role of Ras‐related C3 botulinum toxin substrate 1 (Rac1). Biochem Pharmacol 95: 301‐310, 2015.
 252.Smrcka AV. G protein betagamma subunits: Central mediators of G protein‐coupled receptor signaling. Cell Mol Life Sci 65: 2191‐2214, 2008.
 253.Smrcka AV, Fisher I. G protein betagamma subunits as multi‐functional scaffolds and transducers in G protein‐coupled receptor signaling. Cell Mol Life Sci, 2019. DOI: 10.1007/s00018‐019‐03275‐2.
 254.Snaar‐Jagalska BE, Cambi A, Schmidt T, de Keijzer S. Single‐molecule imaging technique to study the dynamic regulation of GPCR function at the plasma membrane. Methods Enzymol 521: 47‐67, 2013.
 255.Soares GM, Zangerolamo L, Azevedo EG, Costa‐Junior JM, Carneiro EM, Saad ST, Boschero AC, Barbosa‐Sampaio HC. Whole body ARHGAP21 reduction improves glucose homeostasis in high‐fat diet obese mice. J Cell Physiol 233: 7112‐7119, 2018.
 256.Spiegel AM. Signal transduction by guanine nucleotide binding proteins. Mol Cell Endocrinol 49: 1‐16, 1987.
 257.Srivastava M, Eidelman O, Leighton X, Glasman M, Goping G, Pollard HB. Anx7 is required for nutritional control of gene expression in mouse pancreatic islets of Langerhans. Mol Med 8: 781‐797, 2002.
 258.Stan RV, Roberts WG, Predescu D, Ihida K, Saucan L, Ghitescu L, Palade GE. Immunoisolation and partial characterization of endothelial plasmalemmal vesicles (caveolae). Mol Biol Cell 8: 595‐605, 1997.
 259.Stewart A, Fisher RA. Introduction: G protein‐coupled receptors and RGS proteins. Prog Mol Biol Transl Sci 133: 1‐11, 2015.
 260.Stewart M. Molecular mechanism of the nuclear protein import cycle. Nat Rev Mol Cell Biol 8: 195‐208, 2007.
 261.Straub SG, James RF, Dunne MJ, Sharp GW. Glucose augmentation of mastoparan‐stimulated insulin secretion in rat and human pancreatic islets. Diabetes 47: 1053‐1057, 1998.
 262.Straub SG, Sharp GW. Glucose‐stimulated signaling pathways in biphasic insulin secretion. Diabetes Metab Res Rev 18: 451‐463, 2002.
 263.Subasinghe W, Syed I, Kowluru A. Phagocyte‐like NADPH oxidase promotes cytokine‐induced mitochondrial dysfunction in pancreatic beta‐cells: Evidence for regulation by Rac1. Am J Physiol Regul Integr Comp Physiol 300: R12‐R20, 2011.
 264.Sun Z, Zhou T, Pan X, Yang Y, Huan Y, Xiao Z, Shen Z, Liu Z. Design, synthesis and biological evaluation of a series of novel GPR40 agonists containing nitrogen heterocyclic rings. Bioorg Med Chem Lett 28: 3050‐3056, 2018.
 265.Syed I, Jayaram B, Subasinghe W, Kowluru A. Tiam1/Rac1 signaling pathway mediates palmitate‐induced, ceramide‐sensitive generation of superoxides and lipid peroxides and the loss of mitochondrial membrane potential in pancreatic beta‐cells. Biochem Pharmacol 80: 874‐883, 2010.
 266.Syed I, Kyathanahalli CN, Jayaram B, Govind S, Rhodes CJ, Kowluru RA, Kowluru A. Increased phagocyte‐like NADPH oxidase and ROS generation in type 2 diabetic ZDF rat and human islets: Role of Rac1‐JNK1/2 signaling pathway in mitochondrial dysregulation in the diabetic islet. Diabetes 60: 2843‐2852, 2011.
 267.Syed I, Kyathanahalli CN, Kowluru A. Phagocyte‐like NADPH oxidase generates ROS in INS 832/13 cells and rat islets: Role of protein prenylation. Am J Physiol Regul Integr Comp Physiol 300: R756‐R762, 2011.
 268.Syed I, Szulc ZM, Ogretmen B, Kowluru A. L‐threo‐C6‐pyridinium‐ceramide bromide, a novel cationic ceramide, induces NADPH oxidase activation, mitochondrial dysfunction and loss in cell viability in INS 832/13 beta‐cells. Cell Physiol Biochem 30: 1051‐1058, 2012.
 269.Syeda K, Mohammed AM, Arora DK, Kowluru A. Glucotoxic conditions induce endoplasmic reticulum stress to cause caspase 3 mediated lamin B degradation in pancreatic beta‐cells: Protection by nifedipine. Biochem Pharmacol 86: 1338‐1346, 2013.
 270.Tan AM, Samad OA, Fischer TZ, Zhao P, Persson AK, Waxman SG. Maladaptive dendritic spine remodeling contributes to diabetic neuropathic pain. J Neurosci 32: 6795‐6807, 2012.
 271.Tang G, Wang Y, Park S, Bajpayee NS, Vi D, Nagaoka Y, Birnbaumer L, Jiang M. Go2 G protein mediates galanin inhibitory effects on insulin release from pancreatic beta cells. Proc Natl Acad Sci U S A 109: 2636‐2641, 2012.
 272.Tannous M, Veluthakal R, Amin R, Kowluru A. IL‐1beta‐induced nitric oxide release from insulin‐secreting beta‐cells: Further evidence for the involvement of GTP‐binding proteins. Diabetes Metab 28: 3S78‐3S84; discussion 73S108‐73S112, 2002.
 273.Tengholm A, Gylfe E. cAMP signalling in insulin and glucagon secretion. Diabetes Obes Metab 19 (Suppl 1): 42‐53, 2017.
 274.Thamilselvan V, Kowluru A. Paradoxical regulation of glucose‐induced Rac1 activation and insulin secretion by RhoGDIbeta in pancreatic beta‐cells. Small GTPases: 1‐8, 2019. DOI: 10.1080/21541248.2019.1635403.
 275.Tomas A, Yermen B, Min L, Pessin JE, Halban PA. Regulation of pancreatic beta‐cell insulin secretion by actin cytoskeleton remodelling: Role of gelsolin and cooperation with the MAPK signalling pathway. J Cell Sci 119: 2156‐2167, 2006.
 276.Valdivia A, Duran C, San Martin A. The role of Nox‐mediated oxidation in the regulation of cytoskeletal dynamics. Curr Pharm Des 21: 6009‐6022, 2015.
 277.Vasiliauskaité‐Brooks I, Healey RD, Granier S. 7TM proteins are not necessarily GPCRs. Mol Cell Endocrinol 491: 110397, 2019.
 278.Veluthakal R, Chepurny OG, Leech CA, Schwede F, Holz GG, Thurmond DC. Restoration of glucose‐stimulated Cdc42‐Pak1 activation and insulin secretion by a selective Epac activator in type 2 diabetic human islets. Diabetes 67: 1999‐2011, 2018.
 279.Veluthakal R, Chvyrkova I, Tannous M, McDonald P, Amin R, Hadden T, Thurmond DC, Quon MJ, Kowluru A. Essential role for membrane lipid rafts in interleukin‐1beta‐induced nitric oxide release from insulin‐secreting cells: Potential regulation by caveolin‐1+. Diabetes 54: 2576‐2585, 2005.
 280.Veluthakal R, Kaur H, Goalstone M, Kowluru A. Dominant‐negative alpha‐subunit of farnesyl‐ and geranyltransferase inhibits glucose‐stimulated, but not KCl‐stimulated, insulin secretion in INS 832/13 cells. Diabetes 56: 204‐210, 2007.
 281.Veluthakal R, Kumar B, Mohammad G, Kowluru A, Kowluru RA. Tiam1‐Rac1 axis promotes activation of p38 MAP kinase in the development of diabetic retinopathy: Evidence for a requisite role for protein palmitoylation. Cell Physiol Biochem 36: 208‐220, 2015.
 282.Veluthakal R, Madathilparambil SV, McDonald P, Olson LK, Kowluru A. Regulatory roles for Tiam1, a guanine nucleotide exchange factor for Rac1, in glucose‐stimulated insulin secretion in pancreatic beta‐cells. Biochem Pharmacol 77: 101‐113, 2009.
 283.Veluthakal R, Sidarala V, Kowluru A. NSC23766, a known inhibitor of Tiam1‐Rac1 signaling module, prevents the onset of type 1 diabetes in the NOD mouse model. Cell Physiol Biochem 39: 760‐767, 2016.
 284.Veluthakal R, Tunduguru R, Arora DK, Sidarala V, Syeda K, Vlaar CP, Thurmond DC, Kowluru A. VAV2, a guanine nucleotide exchange factor for Rac1, regulates glucose‐stimulated insulin secretion in pancreatic beta cells. Diabetologia 58: 2573‐2581, 2015.
 285.Vikman J, Ahren B. Inhibitory effect of kisspeptins on insulin secretion from isolated mouse islets. Diabetes Obes Metab 11 (Suppl 4): 197‐201, 2009.
 286.Villa SR, Priyadarshini M, Fuller MH, Bhardwaj T, Brodsky MR, Angueira AR, Mosser RE, Carboneau BA, Tersey SA, Mancebo H, Gilchrist A, Mirmira RG, Gannon M, Layden BT. Loss of free fatty acid receptor 2 leads to impaired islet mass and beta cell survival. Sci Rep 6: 28159, 2016.
 287.Villar VA, Cuevas S, Zheng X, Jose PA. Localization and signaling of GPCRs in lipid rafts. Methods Cell Biol 132: 3‐23, 2016.
 288.Villasenor A, Wang ZV, Rivera LB, Ocal O, Asterholm IW, Scherer PE, Brekken RA, Cleaver O, Wilkie TM. Rgs16 and Rgs8 in embryonic endocrine pancreas and mouse models of diabetes. Dis Model Mech 3: 567‐580, 2010.
 289.Vitale N, Gasman S, Caumont AS, Gensse M, Galas MC, Chasserot‐Golaz S, Bader MF. Insight in the exocytotic process in chromaffin cells: Regulation by trimeric and monomeric G proteins. Biochimie 82: 365‐373, 2000.
 290.Vivot K, Moulle VS, Zarrouki B, Tremblay C, Mancini AD, Maachi H, Ghislain J, Poitout V. The regulator of G protein signaling RGS16 promotes insulin secretion and beta‐cell proliferation in rodent and human islets. Mol Metab 5: 988‐996, 2016.
 291.Wang C, Wang W, Liu Y, Yong M, Yang Y, Zhou H. Rac GTPase activating protein 1 promotes oncogenic progression of epithelial ovarian cancer. Cancer Sci 109: 84‐93, 2018.
 292.Wang HW, Mizuta M, Saitoh Y, Noma K, Ueno H, Nakazato M. Glucagon‐like peptide‐1 and candesartan additively improve glucolipotoxicity in pancreatic beta‐cells. Metabolism 60: 1081‐1089, 2011.
 293.Wang J, Li X, Cheng H, Wang K, Lu W, Wen T. Overexpression of Rho‐GDP‐dissociation inhibitor‐gamma inhibits migration of neural stem cells. J Neurosci Res 91: 1394‐1401, 2013.
 294.Wang Q, Pronin AN, Levay K, Almaca J, Fornoni A, Caicedo A, Slepak VZ. Regulator of G protein signaling Gbeta5‐R7 is a crucial activator of muscarinic M3 receptor‐stimulated insulin secretion. FASEB J 31: 4734‐4744, 2017.
 295.Wang X, Elksnis A, Wikstrom P, Walum E, Welsh N, Carlsson PO. The novel NADPH oxidase 4 selective inhibitor GLX7013114 counteracts human islet cell death in vitro. PLoS One 13: e0204271, 2018.
 296.Wang Y, Park S, Bajpayee NS, Nagaoka Y, Boulay G, Birnbaumer L, Jiang M. Augmented glucose‐induced insulin release in mice lacking G(o2), but not G(o1) or G(i) proteins. Proc Natl Acad Sci U S A 108: 1693‐1698, 2011.
 297.Wang Z, Oh E, Thurmond DC. Glucose‐stimulated Cdc42 signaling is essential for the second phase of insulin secretion. J Biol Chem 282: 9536‐9546, 2007.
 298.Wang Z, Thurmond DC. Mechanisms of biphasic insulin‐granule exocytosis – Roles of the cytoskeleton, small GTPases and SNARE proteins. J Cell Sci 122: 893‐903, 2009.
 299.Wang Z, Thurmond DC. Differential phosphorylation of RhoGDI mediates the distinct cycling of Cdc42 and Rac1 to regulate second‐phase insulin secretion. J Biol Chem 285: 6186‐6197, 2010.
 300.Weaver JR, Grzesik W, Taylor‐Fishwick DA. Inhibition of NADPH oxidase‐1 preserves beta cell function. Diabetologia 58: 113‐121, 2015.
 301.Weaver JR, Holman TR, Imai Y, Jadhav A, Kenyon V, Maloney DJ, Nadler JL, Rai G, Simeonov A, Taylor‐Fishwick DA. Integration of pro‐inflammatory cytokines, 12‐lipoxygenase and NOX‐1 in pancreatic islet beta cell dysfunction. Mol Cell Endocrinol 358: 88‐95, 2012.
 302.Wedegaertner PB, Wilson PT, Bourne HR. Lipid modifications of trimeric G proteins. J Biol Chem 270: 503‐506, 1995.
 303.Wennerberg K, Ellerbroek SM, Liu RY, Karnoub AE, Burridge K, Der CJ. RhoG signals in parallel with Rac1 and Cdc42. J Biol Chem 277: 47810‐47817, 2002.
 304.Wertheimer E, Gutierrez‐Uzquiza A, Rosemblit C, Lopez‐Haber C, Sosa MS, Kazanietz MG. Rac signaling in breast cancer: A tale of GEFs and GAPs. Cell Signal 24: 353‐362, 2012.
 305.White CD, Erdemir HH, Sacks DB. IQGAP1 and its binding proteins control diverse biological functions. Cell Signal 24: 826‐834, 2012.
 306.Wieland T. Interaction of nucleoside diphosphate kinase B with heterotrimeric G protein betagamma dimers: Consequences on G protein activation and stability. Naunyn Schmiedebergs Arch Pharmacol 374: 373‐383, 2007.
 307.Wieland T, Nurnberg B, Ulibarri I, Kaldenberg‐Stasch S, Schultz G, Jakobs KH. Guanine nucleotide‐specific phosphate transfer by guanine nucleotide‐binding regulatory protein beta‐subunits. Characterization of the phosphorylated amino acid. J Biol Chem 268: 18111‐18118, 1993.
 308.Wieland T, Ulibarri I, Gierschik P, Jakobs KH. Activation of signal‐transducing guanine‐nucleotide‐binding regulatory proteins by guanosine 5′‐[gamma‐thio]triphosphate. Information transfer by intermediately thiophosphorylated beta gamma subunits. Eur J Biochem 196: 707‐716, 1991.
 309.Willard FS, Crouch MF. Nuclear and cytoskeletal translocation and localization of heterotrimeric G proteins. Immunol Cell Biol 78: 387‐394, 2000.
 310.Wollheim CB, Maechler P. Beta‐cell mitochondria and insulin secretion: Messenger role of nucleotides and metabolites. Diabetes 51 (Suppl 1): S37‐S42, 2002.
 311.Woodard GE, Jardin I, Berna‐Erro A, Salido GM, Rosado JA. Regulators of G protein‐signaling proteins: Negative modulators of G protein‐coupled receptor signaling. Int Rev Cell Mol Biol 317: 97‐183, 2015.
 312.Xia F, Dohi T, Martin NM, Raskett CM, Liu Q, Altieri DC. Essential role of the small GTPase Ran in postnatal pancreatic islet development. PLoS One 6: e27879, 2011.
 313.Xie T, Chen M, Weinstein LS. Pancreas‐specific Gsalpha deficiency has divergent effects on pancreatic alpha‐ and beta‐cell proliferation. J Endocrinol 206: 261‐269, 2010.
 314.Xu J, Zhou X, Wang J, Li Z, Kong X, Qian J, Hu Y, Fang JY. RhoGAPs attenuate cell proliferation by direct interaction with p53 tetramerization domain. Cell Rep 3: 1526‐1538, 2013.
 315.Yaekura K, Julyan R, Wicksteed BL, Hays LB, Alarcon C, Sommers S, Poitout V, Baskin DG, Wang Y, Philipson LH, Rhodes CJ. Insulin secretory deficiency and glucose intolerance in Rab3A null mice. J Biol Chem 278: 9715‐9721, 2003.
 316.Yamaoka M, Ishizaki T, Kimura T. Interplay between Rab27a effectors in pancreatic beta‐cells. World J Diabetes 6: 508‐516, 2015.
 317.Yan J, Yang Y, Zhang H, King C, Kan HM, Cai Y, Yuan CX, Bloom GS, Hua X. Menin interacts with IQGAP1 to enhance intercellular adhesion of beta‐cells. Oncogene 28: 973‐982, 2009.
 318.Yang H, Yang L. Targeting cAMP/PKA pathway for glycemic control and type 2 diabetes therapy. J Mol Endocrinol 57: R93‐R108, 2016.
 319.Yang JY, Walicki J, Abderrahmani A, Cornu M, Waeber G, Thorens B, Widmann C. Expression of an uncleavable N‐terminal RasGAP fragment in insulin‐secreting cells increases their resistance toward apoptotic stimuli without affecting their glucose‐induced insulin secretion. J Biol Chem 280: 32835‐32842, 2005.
 320.Yermen B, Tomas A, Halban PA. Pro‐survival role of gelsolin in mouse beta‐cells. Diabetes 56: 80‐87, 2007.
 321.Yim YY, Zurawski Z, Hamm H. GPCR regulation of secretion. Pharmacol Ther 192: 124‐140, 2018.
 322.Zhang FL, Casey PJ. Protein prenylation: Molecular mechanisms and functional consequences. Annu Rev Biochem 65: 241‐269, 1996.
 323.Zhang P, Kofron CM, Mende U. Heterotrimeric G protein‐mediated signaling and its non‐canonical regulation in the heart. Life Sci 129: 35‐41, 2015.
 324.Zhang X, Damacharla D, Ma D, Qi Y, Tagett R, Draghici S, Kowluru A, Yi Z. Quantitative proteomics reveals novel protein interaction partners of PP2A catalytic subunit in pancreatic beta‐cells. Mol Cell Endocrinol 424: 1‐11, 2016.
 325.Zummo FP, Cullen KS, Honkanen‐Scott M, Shaw JAM, Lovat PE, Arden C. Glucagon‐like peptide 1 protects pancreatic beta‐cells from death by increasing autophagic flux and restoring lysosomal function. Diabetes 66: 1272‐1285, 2017.

Teaching Material

Anjaneyulu Kowluru. GPCRs, G Proteins, and Their Impact on ß-cell Function. Compr Physiol 10 : 2020, 453-490.

Didactic Synopsis

Major Teaching Points:

* A large number of GPCRs are expressed in rodent islets and human islets, and they communicate with trimeric G proteins with high degree of specificity.

* In addition to GPCR-mediated regulation, G proteins are activated via non-canonical mechanisms.

* Activation of GPCR-G protein modules leads to regulation of specific effector proteins leading to the generation of appropriate second messengers, which are essential for physiological insulin secretion.

* GPCR-G protein signaling pathways also regulate functions of monomeric G proteins, which play essential roles in cytoskeletal remodeling and vesicular fusion and insulin exocytosis.

* (In)activation of trimeric and monomeric G proteins is facilitated by highly specific regulatory proteins/factors as well as by specific post-translational modifications.

* Metabolic stress promotes aberrant activation and inappropriate localization of G proteins leading to cell dysfunction and demise.

* Agonists of GPCRs afford protection against β-cell dysfunction observed in in vitro and in vivo models of metabolic stress and diabetes.

Didactic Legends

The following legends to the figures that appear throughout the article are written to be useful for teaching.

Figure 1. Teaching points:

* Ligand binding to a GPCR results in conformational change and activation of a heterotrimeric G protein.

* Several classes of trimeric G proteins have been identified in the islet β-cell with a high degree of specificity for regulation of effector proteins.

* Both α.GTP and βγ activate their respective effector molecules leading to activation or inactivation of cellular signaling pathways.

Figure 2. Teaching points:

* Under basal conditions, α.GDPβγ remains as an inactive complex.

* Once activated α.GTP dissociates from βγ for effector regulation.

* Following functional regulation of respective effectors, the α.GDP re-associates with βγ to attain the inactive conformation.

Figure 3. Teaching points:

* Exposure of pancreatic β-cells to stimulatory glucose leads to the activation of small G proteins, which, in turn, promote a variety of intracellular events including actin cytoskeletal remodeling, transport of insulin granule to the plasma membrane for docking and fusion for release of insulin into the circulation.

Figure 4. Teaching points:

* In a manner akin to trimeric G proteins, small G proteins also undergo activation-inactivation cycles.

* The GTP-bound form of the α-subunit is active and the GDP-bound form is inactive.

* Activation-deactivation cycle is mediated by a variety of regulatory proteins/factors, including GEFs, GAPs and GDIs.

* Some of these regulatory factors are highlighted in Figure 5.

Figure 5. Teaching points:

* This is a logical extension to Figure 4. Several regulatory factors, namely GEFs, GAPs and GDIs precisely control small G protein activation in the islet β-cell.

* These proteins/factors have been implicated in the function of the β-cell, including cell proliferation and physiological insulin secretion.

* In addition to these, relatively less-studied factors are included (under "others") in this Figure.

Figure 6. Teaching points:

* In contrast to classical GPCR-mediated activation of G proteins, published evidence also suggests novel modes of activation of these G proteins via non-canonical mechanisms

* Some of these include regulation by NDPKs, which transiently phosphorylate the β-subunit at a histidine residue; this phosphate, in turn, is relayed to the α.GDP to yield functionally-active Gα.GTP leading to regulation (activation/inactivation) of effector proteins.

Figure 7. Teaching points:

* This figure, which is a logical extension to Figure 6, illustrates the importance of several non-canonical regulators of G protein activation in the islet β-cell.

* GTP/GTP exchange is mediated via multiple proteins/factors, including NDPKs, AGS, GEMS and others.

* Hydrolysis of GTP-bound to α-subunit is mediated via GTPase activity, which is intrinsic to the α-subunit. In addition, RGS proteins accelerate the GTPase activity, thereby terminating the GPCR-G protein coupling and effector activation.

Figure 8. Teaching points:

* The majority of small G proteins, the γ-subunits of trimeric G proteins and nuclear lamins undergo post-translational modifications (prenylation and carboxylmethylation) at their C-terminal cysteine residues.

* These modifications increase the hydrophobicity of the proteins and aid in optimal association with membranes (e.g., targeting) and regulation of effector proteins.

* Available evidence implicates key regulatory roles for these modifications in physiological insulin secretion.

Figure 9. Teaching points:

* Glucose-induced insulin secretion involves activation of a wide-variety of signaling proteins, including PLases. Activation of these enzymes results in the generation of second messenger molecules, including inositol triphosphates and DAG; these are essential for calcium mobilization, cytoskeletal remodeling, and transport of secretory granules.

* Glucose metabolism leads to activation of NDPKs/HK enzymes, which are also implicated in G protein activation.

* Lastly, glucose metabolism results in activation of various GEFs, which promote GTP/GDP exchange on specific G proteins (e.g., Rac1 and Cdc42), which are essential for GSIS.

Figure 10. Teaching points:

* Under physiological conditions, glucose metabolism leads to a tonic increase in intracellular ROS, which, in turn, regulates cytoskeletal remodeling and activation of signaling pathways relevant for insulin secretion.

* In contrast, exposure of pancreatic β-cells to metabolic stress results in increased ROS generation, intracellular oxidative stress and stress kinase activation leading to cell apoptosis.

* Several studies have implicated Rac1 in the regulation of ROS levels under physiological and pathological conditions.

* Such "friendly" and "non-friendly" roles of Rac1 may, in part, be due to its ability to activate Nox2, which is one of the major sources of cellular ROS.

Figure 11. Teaching points:

* This figure highlights the overall impact of GPCR-G protein-effector signalome on islet β-cell function, including GSIS.

* Both canonical and non-canonical modes of activation of G proteins are included herein.

* Activation of these pathways leads to generation of second messenger molecules necessary for cytoskeletal remodeling, vesicular transport and their fusion with the plasma membrane for exocytotic secretion of insulin.

 

* Note that abnormalities in these pathways including GPCR aggregation and/or alterations in post-translational modifications of the candidate proteins in the signalome could lead to dysfunction and demise of the islet β-cell.


Related Articles:

Regulation of G Protein–Coupled Receptors
G Protein‐Coupled Receptors and the G Protein Family
Mediation of Secretory Cell Function by G Protein—Coupled Receptors
Phospholipid‐Derived Second Messengers
Teaching Material

Contact Editor

Submit a note to the editor about this article by filling in the form below.

* Required Field

How to Cite

Anjaneyulu Kowluru. GPCRs, G Proteins, and Their Impact on β‐cell Function. Compr Physiol 2020, 10: 453-490. doi: 10.1002/cphy.c190028