Skip to main content
The Journal of Pharmacology and Experimental Therapeutics logoLink to The Journal of Pharmacology and Experimental Therapeutics
. 2015 May;353(2):235–245. doi: 10.1124/jpet.115.222695

Activators of G Protein Signaling in the Kidney

Frank Park 1,
PMCID: PMC4407716  PMID: 25628392

Abstract

Heterotrimeric G proteins play a crucial role in regulating signal processing to maintain normal cellular homeostasis, and subtle perturbations in its activity can potentially lead to the pathogenesis of renal disorders or diseases. Cell-surface receptors and accessory proteins, which normally modify and organize the coupling of individual G protein subunits, contribute to the regulation of heterotrimeric G protein activity and their convergence and/or divergence of downstream signaling initiated by effector systems. Activators of G protein signaling (AGS) are a family of accessory proteins that intervene at multiple distinct points during the activation–inactivation cycle of G proteins, even in the absence of receptor stimulation. Perturbations in the expression of individual AGS proteins have been reported to modulate signal transduction pathways in a wide array of diseases and disorders within the brain, heart, immune system, and more recently, the kidney. This review will provide an overview of the expression profile, localization, and putative biologic role of the AGS family in the context of normal and diseased states of the kidney.

Introduction

The heterotrimeric G protein signaling system enables the kidney to readily adapt to an ever-changing environment by controlling cellular responses to various hormones and physical stimuli to maintain normal homeostasis of kidney function. The diversity of the heterotrimeric G proteins allows these distinct subunits to perform unique modes of action within the cell, including the mobilization of specific signaling molecules to intracellular organelles or microdomains, receptor phosphorylation and trafficking, and integration of signaling pathways to optimize specificity and/or intensity (Wettschureck and Offermanns, 2005). Historically, canonical G protein signaling involves the stimulation of cell surface G protein–coupled receptors (GPCRs), which function as guanine nucleotide exchange factors (GEFs) to facilitate Gα subunits to release GDP and promote GTP binding. As a consequence of the central role of cell-surface receptors in mediating G protein signaling, there has been enormous interest in the development of drugs to control the signaling output from this GPCR–G protein complex (Wettschureck and Offermanns, 2005; Brogi et al., 2014).

Over the past 20 years, however, the regulation of G protein signaling has become increasingly more diverse through the identification of an increasing number of accessory proteins. Initially, numerous groups discovered negative regulators of GPCR signaling, known as regulators of G protein signaling (RGS) (De Vries et al., 1995; Berman et al., 1996; Druey et al., 1996; Hunt et al., 1996; Koelle and Horvitz, 1996; Siderovski et al., 1996; Watson et al., 1996), which function as GTPase-activating proteins (GAPs) by accelerating the intrinsic GTP catalysis in G protein α-subunits. Subsequently, Cismowski et al. (1999) identified a novel family of distinct GPCR-independent regulators, known as activators of G protein signaling (AGS), using a pheromone receptor–deficient yeast screen. The AGS family members are a compilation of previously unrelated proteins to be grouped together based on protein structure and/or biologic function (i.e., modulate G protein signaling).

Unlike RGS proteins, AGS proteins regulate G proteins with a broader array of mechanisms by forming complexes with Gα or Gβγ independent of the typical heterotrimeric Gαβγ, and are categorized into four separate groups: GEFs, guanine nucleotide dissociation inhibitors (GDIs), Gβγ binding proteins, and Gα16 binding proteins. Although these modes of action by the AGS proteins have been extensively reviewed elsewhere (Sato et al., 2006a; Blumer et al., 2007; Blumer and Lanier, 2014), the biologic function in the kidney remains to be fully determined.

In the normal kidney, AGS proteins appear to be fairly quiescent and play a minimal role in the maintenance of renal function. However, there is emerging and compelling evidence that some of the AGS proteins can play a pivotal role during renal pathologies due to biologic or genetic stress (Nadella et al., 2010; Zheng et al., 2010; Regner et al., 2011; Kwon et al., 2012; Lenarczyk et al., 2014a,b; Potla et al., 2014; Wang et al., 2015). These functional changes in the renal phenotype are likely associated with the ability of AGS proteins to intervene at alternate sites during the typical activation/inactivation cycle of the heterotrimeric G protein complex, as well as the distinct distribution pattern of AGS proteins and their associated heterotrimeric G protein subunits within the nephron, glomerulus, and vasculature.

In this minireview, we will summarize the expression and spatial localization of G proteins and the AGS protein family in normal and pathologic diseased kidneys, and provide some biologic insight into their functional roles in the context of the kidney.

G Protein Subunit Localization in the Kidney

Currently, there are four distinct isoforms of Gα, specifically Gαs, Gαi/o, Gαq/11, and Gα12/13, as well as 5 Gβ and 12 Gγ subunits (Wettschureck and Offermanns, 2005), and their localization can be segment specific within the nephron and renal vasculature. A summary of this section on G protein subunit localization is shown in Table 1.

TABLE 1 .

G protein localization in the kidney

Summary of the vascular and nephron segments that have been identified for Gα subunits in the kidney. Techniques used to determine the presence of the Gα subunits are indicated with lettered footnotes.

Gα Subunit Glomerulus Proximal Convoluted Tubule Thick Ascending Limb Collecting Duct Preglomerular Blood Vessels
Gs +a,b +a,c +c,d +c,d +b
Gi1 +b c d, +c d +b,d
Gi2 +b,c c +d, −c +c,d +b
Gi3 +b,c +c d, +c d +b
Go +b N/D d d N/D
Gq +b − (+ through its interaction with GPCR) d d +b
G11 +b N/D d +d +b
G14 N/D N/D d +d N/D
G15/16 N/D N/D N/D N/D N/D
G12 +c +c +c +c +c
G13 c +c c +c +c

+, positive; −, negative; N/D, not determined.

a

In situ hybridization.

b

Western blot analysis of isolated segments.

c

Immunohistochemistry.

d

Microdissection of renal tubular segments followed by RT-PCR.

Gαs Subunit.

Biodistribution of Gα subunits in human tissues using Northern blot analysis demonstrated minimal to moderate mRNA expression of Gαs compared with Gαi (Brann et al., 1987), which was consistent with the expression of Gαs protein in the whole renal cortex and inner medulla from mice (Yu et al., 1998). Gαs mRNA is widely expressed throughout the renal cells in the nephron and blood vessels. Gαs mRNA was detected in isolated rat nephron segments, including medullary thick ascending limbs and collecting ducts using reverse-transcription polymerase chain reaction (RT-PCR) (Senkfor et al., 1993), and mouse glomeruli and proximal tubules using in situ hybridization (Williamson et al., 1996; Yu et al., 1998). At the protein level, Gαs was detected in the glomeruli (Brunskill et al., 1991; Yanagisawa et al., 1993, 1994; Nitta et al., 1994), proximal convoluted tubules (Brunskill et al., 1991; Stow et al., 1991), thick ascending limbs of Henle (Stow et al., 1991), collecting ducts (Stow et al., 1991), and preglomerular vessels from rats (Ruan et al., 1999).

Gαi/o Subunits.

Brann et al. (1987) demonstrated Gαi2 mRNA expression, but Gαi1 mRNA was undetectable in rat kidneys using Northern blot analysis. However, Hansen et al. (2003) detected all three isoforms of Gαi mRNA in the renal cortex using RT-PCR. To obtain more discrete information on the localization of Gαi subunits within the kidney, RT-PCR analysis was performed on isolated nephron and blood vessel segments from rats (Senkfor et al., 1993; Hansen et al., 2003). Gαi2 was highly identified as the predominant subunit in the medullary thick ascending limb and collecting ducts (Senkfor et al., 1993). Gαi1, Gαi3, and Gαo were undetectable in the nephron segments (Senkfor et al., 1993), but Gαi1 was robustly expressed in preglomerular vessels (Hansen et al., 2003).

Localization of Gαi subunits at the protein level was fairly consistent with spatial expression detected with mRNA assays. Gαi2/i3 was detected in the rat glomeruli using immunohistochemistry (Brunskill et al., 1991; Yanagisawa et al., 1993, 1994) and all Gi1/i2/i3 isoforms were detected in isolated glomerular membranes (Nitta et al., 1994) and preglomerular blood vessels from rats using immunoblot analysis (Ruan et al., 1999). Using immunohistochemistry with rat kidneys, Gαi subunits were segment specific within the nephron, where Gαi1 was localized to the thick ascending limb, including the macula densa, and papillary epithelial cells; Gαi2 was found in the collecting duct cells; and Gαi3 was in the S1 segment proximal tubule cells and macula densa cells in the thick ascending limb of Henle (Stow et al., 1991). Any differences between mRNA and protein localization of the Gαi subunits may be attributed to the relative nonselectivity of the antibodies compared with the nucleotide probes/primers used in the previous studies. In most cases, however, the mRNA and protein results provided confirmatory evidence that one, if not all, of the isoforms exist within specific cell types in the kidney.

Gαo mRNA expression was undetectable in rodent kidneys using Northern blot analysis (Brann et al., 1987; Strathmann et al., 1990) and RT-PCR analyses (Hansen et al., 2003), although RT-PCR analysis showed that a specific GαoA mRNA isoform could be minimally detected in the mouse kidney (Strathmann et al., 1990). Gαo protein expression was minimally expressed in isolated glomerular membranes from humans (Nitta et al., 1994).

Gαq Subunits.

The Gq class of α-subunits consists of Gαq, Gα11, Gα14, and Gα15 (or Gα16 in humans). Gαq, Gα11, and Gα14 mRNA were highly expressed (Strathmann et al., 1989; Wilkie et al., 1991), but Gα15 mRNA was minimally expressed using Northern blot analysis in mouse kidney tissue (Wilkie et al., 1991). Antibodies targeted to Gαq and Gα11 subunits confirmed their expression in isolated whole rat kidney membranes (Gutowski et al., 1991). In human tissue, Gαq presented variable renal mRNA levels from fetus to adult, whereas Gα14 mRNA was only detected in fetal human kidney tissue (Rubio et al., 1999).

Using RT-PCR in isolated rat nephron segments, Gα11 and Gα14 were minimally detected in the outer medullary and upper portion of the inner medullary collecting ducts, respectively (Senkfor et al., 1993). Immunoblot analyses demonstrated Gαq/11 protein in preglomerular vessels (Ruan et al., 1999) and glomeruli from rats (Brunskill et al., 1991; Yanagisawa et al., 1993, 1994).

Gα12/13 Subunits.

Gα12 was robustly detected in the brush border membrane of the proximal tubule, with moderate staining in the thick ascending limb of Henle, cortical collecting ducts, and renal blood vessels in rats (Zheng et al., 2003). In mice, Gα12 was detected uniformly throughout the nephron and the podocytes of the glomeruli (Boucher et al., 2012). Gα13 was similarly expressed in high abundance in the proximal tubules, juxtaglomerular cells, and renal artery with lesser staining in the distal convoluted tubules, medullary collecting ducts, and renal veins (Zheng et al., 2003).

Gβγ Subunits.

The molecular expression of all five Gβ mRNA isoforms was detected in either mouse, chicken, cow, or human kidneys using RNA detection methods. Bovine Gβ1 mRNA (Fong et al., 1987), chicken Gβ3 mRNA (Tummala et al., 2011), and human Gβ5 mRNA (Snow et al., 1999) were detected in the kidneys using Northern blot methods. Mouse Gβ4 (von Weizsäcker et al., 1992) or bovine Gβ2 mRNA (Fong et al., 1987) was not detected in the kidney using Northern blot analysis, but a more sensitive RT-PCR assay followed by Southern blotting of the PCR bands enabled the detection of mouse kidney Gβ1, Gβ2, and Gβ4 mRNA (von Weizsäcker et al., 1992). These contrasting results are likely attributed to the sensitivity of the RNA detection methods, and the lack of bovine Gβ2 mRNA expression in the kidney may be attributed to differential species-specific gene expression regulation.

Of the 12 Gγ isoforms, only 6 were detected in the kidney. Northern blot analysis was used to detect the mRNA for human Gγ4 (Ray et al., 1995), bovine Gγ5 (Cali et al., 1992), bovine Gγ7 (Cali et al., 1992), human Gγ10 (Ray et al., 1995), and human Gγ11 (Ray et al., 1995). Gγ12 protein was detected in the rat kidneys using Western blot analysis (Asano et al., 1998).

The spatial localization of each distinct β or γ isoforms within distinct cell types in the kidney remains to be determined. However, the diversity of each distinct Gα subunit within specific cell types of the kidney may be more expansive than described using molecular techniques, particularly for Gαq. Many published studies investigating GPCR activity have described Gα function to specific vascular, tubular, or interstitial cells within the kidney, without actual evidence for the presence of those G protein subunits being expressed in those segments (Zhang et al., 2011; Zhuo and Li, 2011; Horita et al., 2013). As an example, angiotensin II is well known to regulate proximal tubular sodium reabsorption through activation of the AT1A receptor and its association with Gαq, but there are no definitive data showing the presence of this subunit in that particular segment of the kidney other than interaction studies unrelated to the kidney (Sano et al., 1997; Li et al., 2011b).

Regardless, these molecular studies describing the expression of Gα, Gβ, and Gγ subunits in the kidney provides some potential insight into the types of interactions that may occur with accessory proteins in each respective renal cell type, but further studies are needed to confirm the identity of particular G protein subunits within specific segments of the kidney.

AGS Proteins

Since the initial findings by Cismowski et al. (1999) using the genetically modified yeast system, there are a total of 19 accessory proteins divided into 4 distinct groups within the AGS family. Thirteen AGS proteins were identified using cDNA libraries from multiple organ systems, and an additional 6 accessory proteins were added based on their mode of action (Sato et al., 2006a; Blumer et al., 2007). The renal localization of the 13 AGS proteins identified using the yeast screen were summarized in Table 2.

TABLE 2.

Localization of AGS proteins in the kidney

Summary of the localization of AGS family members within distinct segments of the normal kidney using immunohistochemistry. In studies with low staining, the cell types could not be determined.

Protein Alternate Name Glomerulus Proximal Convoluted Tubule Thick Ascending Limb Collecting Duct Preglomerular Blood Vessels
AGS1 RasD1/Dexras1 +
AGS2 Tctex1 N/D N/D N/D N/D N/D
AGS3 GPSM1 a +
AGS4 GPSM3/G18.1b N/D N/D N/D N/D N/D
AGS5 GPSM2/LGN + +
AGS6 RGS12 + + + + +
AGS7 Trip13/PCH2/16E1-BP +
AGS8 FNDC1/KIAA1866 + +
AGS9 Rpn10/Psmd4 N/D N/D N/D N/D N/D
AGS10 GNAO + N/D N/D N/D N/D
AGS11 TFE3 b b b
AGS12 TFEB b b b
AGS13 MitF N/D N/D N/D N/D N/D

−, low to absent expression; +, detectable protein expression; N/D, not specifically determined.

a

Detectable protein expression after ischemia-reperfusion injury.

b

Low-to-undetectable levels for TFE3 and TFEB in the cytoplasm of renal tubules.

Group I AGS Proteins

There are currently four AGS proteins that are categorized in this group: 1) Activator of G protein Signaling 1 (AGS1)/RasD1/Dexras1; 2) RasD2/Ras homolog enriched in striatum (Rhes)/tumor endothelial marker 2 (TEM2); 3) GIV/Girders of actin filaments (Girdin)/Akt phosphorylation enhancer (APE); and 4) resistance to inhibitors of cholinesterase 8 (Ric-8). The latter three accessory proteins were not identified from the yeast screen, but have similar protein structure and/or mechanism of action so they have been included as part of the group I AGS proteins.

AGS1/Dexras1/RasD1.

AGS1 was the initial AGS protein that was characterized by Cismowski et al. (1999), and demonstrated homology with a dexamethasone-inducible Ras protein, known as Dexras1 or RasD1 (Cismowski et al., 2000). Ras proteins are a family of small (typically 20–25 kDa) monomeric G proteins, which can alternate the binding of guanine nucleotides (GDP and GTP) to control the signaling output within the cell. AGS1 demonstrates selective interaction with Gαi, and not Gαs, Gα16, or Gβγ subunits (Cismowski et al., 1999, 2000, 2001). In yeast, overexpression of AGS1 with either mutant Gαi2 (Cismowski et al., 1999, 2000) or RGS4 (Cismowski et al., 1999), a GAP for Gi/Go proteins, prevented the functional signaling output mediated by AGS1. These studies demonstrate that the biologic function of AGS1/Dexras1 requires the GTP-bound forms of Gαi subunits to activate signaling pathways (Tu and Wu, 1999).

The renal expression of AGS1/Dexras1 mRNA was developmentally regulated where higher levels were detected in the adult versus fetus (Kemppainen et al., 2003). In terms of tissue biodistribution, AGS1/Dexras1 mRNA was minimally expressed in adult mice (Kemppainen and Behrend, 1998) or human kidneys (Tu and Wu, 1999; Kemppainen et al., 2003) compared with other sites, including the liver and brain. AGS1/Dexras1 mRNA expression was induced by dexamethasone administration (Kemppainen and Behrend, 1998; Tu and Wu, 1999; Brogan et al., 2001; Vaidyanathan et al., 2004), including in the kidney (Kemppainen and Behrend, 1998), or by biologic stress applied to the kidney (Lenarczyk et al., 2014a,b). Coincidentally, dexamethasone has been shown to exhibit a protective role in the kidney after renal ischemia-reperfusion injury (Rusai et al., 2013). Considering that AGS1/Dexras1 protein was predominantly localized to the cortical and outer medullary proximal tubular epithelial cells in normal mouse kidneys (Lenarczyk et al., 2014a,b), which are nephron sites that are highly sensitive to ischemia (Devarajan, 2006; Bonventre and Yang, 2011), there may be a role for AGS1/Dexras1 to modulate the extent of renal tubular epithelial cell injury through its ability to activate downstream signaling pathways (Tu and Wu, 1999), which are involved in cell growth, differentiation, and transformation (Kemppainen and Behrend, 1998).

There may be additional roles for AGS1/Dexras1 in the kidney to control cardiovascular function. AGS1/Dexras1 can control renin transcription (Tan et al., 2011) or nitric oxide synthase activity (Fang et al., 2000), which are well established to play a crucial role in long-term sodium reabsorption and blood pressure regulation by controlling regional blood flow distribution and fluid and electrolyte balance in the kidney (O'Connor and Cowley, 2010; Aksu et al., 2011; Toda and Okamura, 2011). In cell culture, AGS1/Dexras1 modulated cAMP production by adenylyl cyclase II through the actions of the dopamine D2 receptor (Nguyen and Watts, 2006). This may implicate another alternate role for AGS1/Dexras1 in the kidney by regulating Na+-K+ ATPase activity in a cAMP-dependent mechanism through the dopamine D2 receptor (Bertorello and Aperia, 1990). The functional roles of AGS1/Dexras1 and its effect on renal function require further investigation.

RasD2/Rhes/TEM2.

RasD2/Rhes/TEM2 has moderate sequence homology (approximately 60%) to AGS1/RasD1. Rhes/RasD2 was highly expressed in the striatum with minimal detection in the kidney using RNA blot analysis (Spano et al., 2004). Unlike AGS1/Dexras1, Rhes/RasD2 has yet to be confirmed as a GEF, so further evaluation is necessary to confirm whether it fulfills the criteria to be categorized as a legitimate AGS protein (Falk et al., 1999; Vargiu et al., 2004; Thapliyal et al., 2008; Harrison and He, 2011).

GIV/Girdin/APE.

GIV/Girdin/APE was initially shown to regulate actin organization and cell motility by acting as a substrate for Akt phosphorylation. Subsequently, studies have demonstrated that the C terminus of GIV/Girdin can directly interact with receptor tyrosine kinases, G protein subunits, and Akt to control processes, including cell migration (Ghosh et al., 2008, 2010), autophagy (Garcia-Marcos et al., 2011), and metastasis (Ghosh et al., 2010). Initial biodistribution studies by Anai et al. (2005) were unable to reliably detect the expression of GIV/Girdin/APE mRNA and protein in the kidney using Northern blot and immunoblot analysis after immunoprecipitation of tissue lysates, respectively. More recently, the expression of GIV/Girdin was highly induced in glomerular podocytes after administration of puromycin aminonucleoside in rats (Wang et al., 2015), which has been previously shown to be an animal model of reversible podocyte injury (Pippin et al., 2009). In this study, GIV/Girdin promoted the formation of a signaling complex with Gαi and vascular endothelial growth factor receptor 2, leading to elevated phosphoinositide-3 kinase/Akt survival signaling and protection from damage by puromycin aminonucleoside (Wang et al., 2015).

Ric-8.

In mammals, there are two isoforms, Ric-8A and Ric-8B, which function as GEFs for the Gα subunits (Tall et al., 2003; Chan et al., 2011). Ric-8A promoted the intrinsic nucleotide exchange rate for Gαi, Gαq, and Gα12/13, but Ric-8B interacts only with Gαs subunits. Ric-8A functions as a chaperone protein to direct Gα subunit association to the plasma membrane. In the absence of Ric-8A/Ric-8B, the steady state levels of Gα are appreciably reduced suggesting a role for Ric-8 to stabilize the Gα subunit from degradation during the biosynthetic process (Gabay et al., 2011). To date, the role of Ric-8 in the kidney remains largely unexplored since Ric-8A was exclusively detected in the nervous system of lower invertebrates and mice (Miller et al., 2000; Tõnissoo et al., 2003, 2006; Maldonado-Agurto et al., 2011). Using a more sensitive RT-PCR assay, however, adult frogs presented a more widespread expression pattern for Ric-8 mRNA, including modest expression in the kidney, but these levels may not be sufficient to alter renal function.

Group II AGS Proteins

Group II AGS proteins are categorized by virtue of one or more protein sequences known as GoLoco (Siderovski et al., 1999) or G protein regulatory (GPR) domains (Peterson et al., 2000; Bernard et al., 2001). Depending on the protein structure, three distinct types of group II AGS proteins have been identified.

AGS3/GPSM1.

AGS3 is also known as G protein signaling modulator 1 (GPSM1), and was first described by Takesono et al. (1999) as the initial member of the GDIs. AGS3/GPSM1 has a protein structure containing N-terminal tetratricopeptide repeats, a linker region, and four C-terminal GPR/GoLoco motifs, which can bind one or more Gαi/o subunits and regulate heterotrimeric G protein activity (Takesono et al., 1999; Peterson et al., 2000; Bernard et al., 2001).

Under basal conditions, full-length and truncated forms of AGS3/GPSM1 mRNA (De Vries et al., 2000a; Pizzinat et al., 2001) and protein (De Vries et al., 2000a; Pizzinat et al., 2001; Blumer et al., 2002; Nadella et al., 2010; Regner et al., 2011; Kwon et al., 2012) were expressed at minimal to undetectable levels in mouse or rat kidneys. This was likely attributed to the selective localization of AGS3/GPSM1 to the distal portion of the nephron (Nadella et al., 2010; Regner et al., 2011). No expression was detected in other nephron segments of the renal cortex or medulla, and no expression was detected in the vasculature or glomerulus.

Under conditions of acute or chronic kidney injury to the renal tubular epithelia, the extent of AGS3/GPSM1 protein induction was dependent upon the site of epithelial cell injury or disruption (Nadella et al., 2010; Regner et al., 2011; Kwon et al., 2012). Ischemia-reperfusion injury, a common form of acute kidney injury, resulted in a dramatic temporal induction of AGS3/GPSM1 expression during the reparative phase of recovery in the outer medullary proximal tubular epithelial cells (Regner et al., 2011; Lenarczyk et al., 2014a,b), which was a nephron segment normally absent in AGS3/GPSM1 expression. In multiple genetic models of polycystic kidney disease from mice, rats, and humans (Nadella et al., 2010; Kwon et al., 2012), AGS3/GPSM1 expression was abnormally elevated in the cystic tubular epithelial cells derived from the collecting ducts (Nadella et al., 2010; Kwon et al., 2012).

However, AGS3/GPSM1 does not appear to play a role during compensatory hypertrophy, which is an alternate mechanism to produce increased cell size (Lenarczyk et al., 2014b). During hypertrophy, AGS3/GPSM1 protein expression was not induced (Lenarczyk et al., 2014b). Moreover, unilateral nephrectomy in the Gpsm1-deficient mice did not affect the ability of the remaining kidney to increase its size compared with wild-type mice (Lenarczyk et al., 2014b).

From these studies, AGS3/GPSM1 appeared to act as a stress-response protein to promote tubular epithelial cell repair in an attempt to restore normal kidney function after renal injury or in genetic diseases involving cellular hyperplasia.

Mechanistically, AGS3/GPSM1 sequesters Gαi subunits to prevent the inactivation of Gβγ signaling (Nadella et al., 2010; Regner et al., 2011; Kwon et al., 2012). Gβγ activity has been implicated in mitotic spindle orientation (Sanada and Tsai, 2005), ion channel activity (Kwon et al., 2012), cAMP production (Fan et al., 2009), or modulation of GPCR-dependent (Sato et al., 2004; Oner et al., 2010, 2013) and GPCR-independent signaling pathways (Smrcka, 2008; Smrcka et al., 2008; Lin and Smrcka, 2011). Overexpression of AGS3/GPSM1 facilitated an increase of polycystin-1/-2 ion channel activity, which could be blocked by a scavenger of Gβγ dimers (Kwon et al., 2012). Alternatively, there was evidence to suggest that AGS3/GPSM1, through its interaction with Gαi subunits, could regulate the autophagic process (Pattingre et al., 2003, 2004; Ghosh et al., 2010; Groves et al., 2010), but this may be cell-type dependent (Vural et al., 2013). AGS3/GPSM1 may also influence signal transduction pathways through other protein–protein interactions, such as liver kinase B1 (Blumer et al., 2003), which is a serine/threonine kinase known to regulate AMP-activated protein kinase signaling during states of energetic stress (Alexander and Walker, 2011). These studies demonstrate the potentially diverse pathways that may be controlled by AGS3/GPSM1 during normal or pathologic states.

AGS5/GPSM2/LGN.

AGS5 is also known as LGN (Leu-Gly-Asn repeat-enriched protein) or G protein signaling modulator 2 (GPSM2), and functions as a GDI. AGS5/LGN exhibits a similar protein structure as AGS3/GPSM1, and is considered a mammalian homolog of AGS3/GPSM1. Unlike AGS3/GPSM1, AGS5/LGN was ubiquitously expressed in mammalian organs, including the kidney (Blumer et al., 2002). Our group has detected AGS5/LGN in mouse, rat, and human tubular epithelial cell lines in vitro and whole kidneys in vivo (Nadella et al., 2010; Regner et al., 2011; Kwon et al., 2012). In the normal rat kidneys, AGS5/LGN was detected in the distal tubules, including thick ascending limb and collecting ducts (Lenarczyk et al., 2014a,b). Unlike AGS3/GPSM1, there was no observable change in the AGS5/LGN protein levels or localization pattern after acute kidney injury (Regner et al., 2011; Lenarczyk et al., 2014b) or in genetically mutant cystic kidneys (Nadella et al., 2010).

AGS5/LGN and AGS3/GPSM1 are also considered as mammalian homologs of the Drosophila Partner of Inscuteable (Pins) protein. Functionally, AGS5/LGN has been well characterized as a regulator of mitotic spindle orientation, which is a process that participates in asymmetric cell division of epithelial cells (Fuja et al., 2004; Lechler and Fuchs, 2005; Culurgioni et al., 2011; Zhu et al., 2011). Consistent with these findings, AGS5/LGN was determined to play a key role in cyst formation by controlling mitotic spindle orientation in a renal epithelial cell system in vitro (Zheng et al., 2010; Xiao et al., 2012), but the LGN-Gαi complex may not be involved in asymmetric daughter cell formation (Xiao et al., 2012). These findings may suggest that AGS5/LGN could compensate for the loss of its homolog, AGS3/GPSM1, as observed in a recent study investigating acute or chronic kidney injury (Regner et al., 2011; Kwon et al., 2012). Alternatively, there may be other biologic roles for AGS5/LGN in tubular epithelial cells in the kidney by virtue of its interaction with other proteins, such as soluble guanylate cyclase (Chauhan et al., 2012), which is an important regulator of fluid and electrolyte balance through the actions of hormones, such as nitric oxide (O'Connor and Cowley, 2010; Aksu et al., 2011; Toda and Okamura, 2011). To date, the loss of AGS5/LGN in the kidney and its effects on renal function remains to be determined.

The second type of group II AGS proteins, including AGS6, RGS14, and Rap1GAP, were identified by the presence of a single GPR motif and other protein regulatory domains that function to accelerate Gα-GTP hydrolysis (Blumer and Lanier, 2014).

AGS6/RGS12.

AGS6 has sequence identity with RGS12, which is the largest member of the RGS protein family. AGS6/RGS12 has a multidomain protein structure that contains the RGS domain, a single GoLoco/GPR motif, and other protein binding domains. AGS6/RGS12 inactivated G protein signaling by accelerating the intrinsic GTPase activity for Gαi/o subunits through the N-terminal RGS domain (De Vries et al., 2000b), whereas the C-terminal GPR/GoLoco motif interacted with all isoforms of the Gαi subunits (Kimple et al., 2001). The expression profile for AGS6/RGS12 in the kidney may be species or isoform dependent, since multiple RGS12 mRNA isoforms were expressed in the human kidneys (Snow et al., 1998), but not in mouse kidneys (Yang and Li, 2007). At the protein level, however, we have observed low uniform staining throughout the rat nephron and the endothelial cells of the blood vessels in the renal cortex (Lenarczyk et al., 2014b).

Functionally, there is a paucity of data regarding the role of AGS6/RGS12 in the kidney, but there are number of studies demonstrating that AGS6/RGS12 played a critical role in the regulation of calcium channels, including N-type (Schiff et al., 2000; Yang and Li, 2007) and CaV2.2 calcium channels (Anantharam and Diversé-Pierluissi, 2002; Richman and Diversé-Pierluissi, 2004; Richman et al., 2005). At this time, the role of the GPR domain in RGS12 and how it regulates G protein signaling compared with its RGS effects remains to be determined.

RGS14.

RGS14 is an accessory protein initially cloned by Snow et al. (1997), and contains a single GPR/GoLoco motif and a RGS domain. RGS14 interacted with Gαi/o subunits in the RGS box (Traver et al., 2000; Kimple et al., 2001), whereas the GPR/GoLoco motif selectively bound to Gαi1 and Gαi3 (Mittal and Linder, 2004) with weaker interaction with Gαi2 (Kimple et al., 2001). RGS14 mRNA distribution was relatively exclusive to the spleen, lung, and select regions in the rat brain (Snow et al., 1997; Reif and Cyster, 2000) with no apparent expression in the kidney (Snow et al., 1997).

Rap1GAP.

Rap1GAP has limited expression distribution in mammalian organs under normal conditions (Rubinfeld et al., 1991). Recent studies, however, have shown that the regulation of Rap1GAP expression was dependent upon the disease stimulus in the kidney. Rap1GAP expression was induced in renal glomerular podocytes from HIV-1 transgenic mice and in human kidney biopsies of focal and segmental glomerulosclerosis (Potla et al., 2014), which appeared to contribute to the podocyte dysfunction after injury by altering β1-integrin–mediated adhesion. In renal carcinoma, the Rap1GAP protein levels were reduced, leading to increased cellular invasion (Kim et al., 2012). The importance of these observations in terms of renal function needs further elucidation.

The third type of group II AGS proteins consists of three proteins: a truncated form of AGS3 (AGS3-SHORT) (Pizzinat et al., 2001), AGS4/GPSM3/G18.1b (Cao et al., 2004), and Pcp-2/L7/GPSM4 (Nordquist et al., 1988), which has multiple C-terminal GPR domains, but no other defined regulatory protein binding sites. Minimal-to-no expression of AGS3-SHORT (Pizzinat et al., 2001), AGS4/GPSM3/G18.1b (Cao et al., 2004; Zhao et al., 2010), or Pcp-2/L7/GPSM4 (Nordquist et al., 1988; Saito et al., 2005) has been detected in the kidneys.

Group III AGS Proteins

Group III AGS proteins include AGS2/TcTex1 (DiBella et al., 2001), AGS7/Trip13 (Li and Schimenti, 2007; Roig et al., 2010), AGS8/FNDC1 (Nielsen et al., 1993), AGS9/Rpn10/S5a/Psmd4 (Deveraux et al., 1995), and AGS10/GNAO (Neer et al., 1984; Sternweis and Robishaw, 1984). The biologic function of group III AGS proteins remains largely undefined in the kidney, but each of these proteins interacts directly with Gβγ subunits complexed with or without the Gα subunits.

AGS2/Tctex1.

AGS2 was initially characterized by Takesono et al. (1999), and was homologous to Tctex1. Tctex1 is a cytoplasmic dynein light chain essential for dynein assembly and participates in specific motor cargo interactions (DiBella et al., 2001). Previous studies have detected Tctex1 mRNA (DiBella et al., 2001) and protein (King et al., 1998) in fetal and adult mouse kidneys (King et al., 1998; DiBella et al., 2001). Human kidneys express higher levels of Tctex1 mRNA compared with the other organs analyzed (DiBella et al., 2001). Recent studies using neural progenitor cells have shown that AGS2/Tctex1 is recruited to the ciliary transition zone prior to the DNA synthesis phase of the cell cycle (Li et al., 2011a), and is modulated by interaction with AGS3/GPSM1 (Yeh et al., 2013). For this reason, AGS2/Tctex1 may have clinical relevance with renal pathologies associated with ciliary defects, such as polycystic kidney disease.

AGS7/Trip13/mPCH2.

AGS7 was isolated from a yeast screen investigating receptor-independent G protein regulators using a prostate leiosarcoma cDNA library (Sato et al., 2006b). AGS7 demonstrated sequence identity with the C-terminal portion of the thyroid receptor–interacting protein 13 (Trip13) (Cismowski and Lanier, 2005), which was originally identified among a group of other proteins that interacted with the thyroid hormone receptor in a yeast two-hybrid screen (Lee et al., 1995). Neither the role of AGS7/Trip13 as a regulator of the thyroid hormone receptor nor G protein signaling functions have been elucidated in the kidney in vivo. Instead, most of the work involving AGS7/Trip13, which is also a homolog to mouse pachytene checkpoint 2 (mPCH2), has focused on its AAA-ATPase activity with emphasis on the mechanistic control of DNA damage repair (Bolcun-Filas et al., 2014), cell-cycle checkpoint, kinetochore control (Tipton et al., 2012; Wang et al., 2014), and meiotic recombination (Li and Schimenti, 2007; Roig et al., 2010). These initial studies led to recent high-impact findings regarding AGS7/Trip13 in reproductive cell biology (Li and Schimenti, 2007; Roig et al., 2010) and cancer (Banerjee et al., 2014).

In the kidney, the expression of Trip13 mRNA was minimally detected in the kidney using qualitative RT-PCR (Li and Schimenti, 2007). This was consistent with the Trip13 protein localization found only in the principal cells of the collecting ducts in the normal rat kidney (Lenarcyzk et al., 2014a). The site of expression in the nephron may implicate AGS7/Trip13 as a potential regulator of Na+, water, or urea reabsorption, but the biologic importance of AGS7/Trip13 during normal and pathologic conditions in the kidney needs further investigation.

AGS8/FNDC1/KIAA1866.

AGS8 was identified using cDNA library from the ischemic injured mouse hearts (Nielsen et al., 1993), and was homologous to Fibronectin type III containing I (FNDC1) (Gao et al., 2003) and KIAA1866 (GenBank accession no. XM_217792.2). Transcript profiling by RT-PCR analysis showed that the basal expression of AGS8 mRNA was most abundant in the skeletal muscle and the kidney (Sato et al., 2006b). AGS8/FNDC1 localization was detected exclusively in distal tubular epithelial cells using immunohistochemistry of Sprague-Dawley rat kidneys (Lenarcyzk et al., 2014a).

Little is known about the function of AGS8/FNDC1 in the kidney. A recent genome-wide association study suggested a potential link between AGS8/FNDC1 and urinary albumin-to-creatinine ratio in African Americans from the CARe Renal Consortium (Liu et al., 2011). However, morpholino knockdown of AGS8/FNDC1 in zebrafish did not support a role for this protein during renal development or dysfunction (Liu et al., 2011).

In terms of G protein regulation, epistasis experiments demonstrated that AGS8 interacted selectively with Gβγ, but not Gαi1/2/3/0 or Gαs. AGS8 has been shown to interact with Gβγ activates connexin-43 (CX-43), which can regulate gap junction function (Sato et al., 2009). Considering that CX-43 is abundantly expressed in the glomerulus and inner medullary collecting duct epithelial cells (Guo et al., 1998), which are a primary site of AGS8 expression, the role of AGS8 with connexins, including CX-43, may need further exploration in the kidney. Alternatively, AGS8/Fndc1 may play an important role in ischemia-reperfusion injury. In the heart, AGS8/Fndc1 gene expression could be robustly induced in cardiomyocytes during ischemia (Sato et al., 2006b, 2009, 2014). In the kidney, however, our laboratory detected a marked reduction in AGS8/Fndc1 mRNA after 24 hours after ischemia-reperfusion injury, which slowly returned toward normal levels after 168 hours (Lenarcyzk et al., 2014a). It is not presently known whether the reduction in renal AGS8/Fndc1 mRNA is attributed directly to negative transcriptional regulation or whether damaged nephron segments were unable to continue expressing AGS8/Fndc1 due to the biologic insult.

AGS9/PSMD4/Rpn10/S5a.

AGS9 has sequence identity to a protein known as S5a, which was encoded by the gene for the multiubiquitin-chain-binding protein (Mcb1) in rodents and humans (Ferrell et al., 1996; Pusch et al., 1998). S5a has multiple other nomenclatures, including Rpn10 and Psmd4. S5a/Rpn10 was ubiquitously expressed, but it has multiple splice variants, Rpn10a, Rpn10b, Rpn10c, Rpn10d, and Rpn10e. The expression pattern was dependent on the stage of development (Hamazaki et al., 2007), and the kidneys were one of the major organs that had the highest level of S5a/Rpn10 expression using Northern blot analysis (Pusch et al., 1998).

S5a is part of a large multimeric protein complex involved in protein degradation through the ubiquitin-proteosome system. In the ubiquitin-proteosome system, protein substrates that were destined for degradation were polyubiquitinated for subsequent catabolism through a nonlysosomal pathway (Ciechanover, 1993), which involved the 26S proteasome (Tanaka, 1995). The binding of the polyubiquitinated protein substrate to the 26S proteasome was facilitated by the S5a subunit, and was one of the major docking sites in the proteosome (Verma et al., 2004). Genetic loss of the S5a/Rpn10 expression in mice resulted in embryonic lethality (Hamazaki et al., 2007), and this would suggest that S5a/Rpn10-mediated degradation of ubiquitinated proteins was an indispensable cellular function essential to maintain mammalian life. To date, it remains to be determined which heterotrimeric G protein subunits, namely Gβγ, interact with S5a/Rpn10 to control renal function.

AGS10/GNAO.

The more commonly known name for AGS10 is GNAO, which is a G protein α subunit that was initially isolated as a 39-kDa polypeptide from rat brain and liver, and bovine heart (Neer et al., 1984; Sternweis and Robishaw, 1984). AGS10/GNAO expression appears to be more broadly detected in many other cell types, including pancreatic cells (El-Mansoury and Morgan, 1998), but there is minimal, if any, expression within the kidney (Brann et al., 1987; Senkfor et al., 1993). Because of a lack of GNAO expression in the kidney as well as the lethality of GNAO knockout mice, there is minimal knowledge regarding any role for GNAO in the kidney.

Group IV AGS Proteins

The most recent AGS proteins were identified by their interaction with a specific subunit, Gα16, in the Gαq family (Sato et al., 2011). Sequence analysis of the Gα16-specific cDNAs were identified as the microphthalmia family (MiTF) of basic helix-loop-helix-leucine zipper transcription factors (Sato et al., 2011), specifically transcription factor E3 (TFE3), transcription factor EB (TFEB), and MiTF. Numerically, TFE3, TFEB and MiTF were designated as AGS11, AGS12, and AGS13, respectively (Sato et al., 2011).

In normal kidneys, the mRNA expression profile of AGS11/TFE3, AGS12/TFEB, and AGS13/MiTF is generally quite low (Kuiper et al., 2003, 2004), and the protein localization is either observed predominantly in the cytoplasm or weakly in the nucleus (Hong et al., 2010). There is some evidence that the basal role of TFE3 in proximal tubules is to control inorganic phosphate reabsorption (Miyamoto and Itho, 2001). However, MitF/TFE transcription factors appears to play a fundamentally important role during translocation renal cell carcinomas from children and young adults (Armah and Parwani, 2010; Hong et al., 2010), in which there is modest to robust nuclear localization of AGS11/TFE3 and to a lesser extent, AGS12/TFEB. Because of this shift in subcellular location to the nucleus, the MiTF/TFE transcription factors are being used as diagnostic biomarkers for specific subtypes of renal cell carcinoma. In these subtypes of renal cancer, the cellular transformation is believed to be attributed to abnormal chromosomal translocation or inversion of genomic DNA containing either TFE3 (Meloni et al., 1993; Weterman et al., 1996a,b) or TFEB (Kuiper et al., 2003) into other genes (Weterman et al., 2000, 2001; Mathur and Samuels, 2007). To date, the nephron segment responsible for the renal cell carcinoma remains to be determined, but there is evidence for TFEB translocation originating from distal tubules (Rao et al., 2012).

MiTF has also been linked as an oncogene for renal cell carcinoma, but its function has been described in greater detail for melanoma (Granter et al., 2002; Bertolotto et al., 2011). There is evidence that MiTF/TFE transcription factors have an inter-relationship between each other through redundant oncogenic function whereby TFE3 knockdown decreased viability of TFE3-translocated papillary renal carcinoma cells, but could be rescued by expression of MiTF (Davis et al., 2006).

In addition to renal oncogenesis, there is increasing evidence that TFE3 plays a role in the pathogenesis of renal cystic disease. Increased chimeric TFE3 protein led to dedifferentiation of renal proximal tubule cells as determined by a loss of cilia formation and key functional transporter proteins (Mathur and Samuels, 2007). Other studies have shown that folliculin, a crucial negative regulator of TFE3 nuclear translocation, promoted renal cell carcinoma and produced the phenotypic appearance of polycystic kidney disease (Chen et al., 2008; Hong et al., 2010). In folliculin-deficient mice, renal tubular epithelial cells transformed into a cystic phenotype that increased nuclear localization of TFE3 compared with the noncystic kidneys (Chen et al., 2008).

There currently remains a paucity of information regarding the mode of action by which MitF/TFE regulates the process of oncogenesis and cystogenesis, particular its role in modulating G protein signaling in the kidney. Gα16 was normally localized to the cytoplasm and plasma membrane (Sato et al., 2011). Coexpression of Gα16 with TFE3 led to nuclear accumulation, which induced the expression of claudin 14, a cell junction protein, in cardiomyocytes (Sato et al., 2011). Claudins are widely expressed in the kidney and could implicate these transcription factors as regulators of paracellular ion transport to control fluid and electrolyte balance (Yu, 2015). On the other hand, MiTF has been shown to be a transcriptional regulator of hypoxia inducible factor 1α (Buscà et al., 2005), which was shown to be abundantly expressed in various renal cell types (Haase, 2006).

There is definitely a need to further elucidate the functional role of these transcription factors with respect to G protein regulation. Since the expression of Gα15/16 is much lower compared with other isoforms of the Gαq isoforms in the kidney (Wilkie et al., 1991), it remains to be determined whether this is a signaling mechanism by which AGS11–AGS13 promote carcinogenesis in the kidney.

Summary and Perspective

AGS proteins exhibit a distinct distribution pattern within the nephron, glomerulus, and vasculature, which helps in producing a diverse control mechanism on G protein signaling evoked by receptor-dependent or receptor-independent stimuli. Although none of the currently known AGS proteins were initially discovered using a normal or diseased kidney-specific cDNA library, several have shown biologic importance in the kidney, including AGS3 during acute and chronic kidney injury (Nadella et al., 2010; Regner et al., 2011; Kwon et al., 2012) and AGS11–AGS13 in different forms of renal cell carcinoma (Granter et al., 2002; Armah and Parwani, 2010; Hong et al., 2010; Bertolotto et al., 2011). Further progress regarding the importance of AGS proteins in the kidney is needed under various stressful conditions, and more importantly, to determine whether the mode of regulation is through the perturbation of G protein signaling. Some of the identified AGS proteins, such as AGS2, AGS7, AGS9, and AGS11–AGS13, were not previously known to interact with Gα or Gβγ subunits, and have only begun to be studied in greater detail.

Identification of additional AGS proteins, if they even exist, using kidney-specific cDNA libraries in the yeast screen may also offer new avenues of research in normal, and possibly diseased, kidneys. Even without the identification of new AGS family members, there will be continual accumulation of knowledge regarding the expression profile, spatial localization, and biologic function of the AGS proteins in various renal pathologies. This will warrant the development of therapeutic interventions targeting individual AGS proteins, particularly in disease states with an active state of growth or hyperplasia. As an example, group II AGS proteins contain one or more of the unique GPR motifs that could be targeted using peptide sequences to manipulate the binding of Gαi subunits in the renal epithelial cells (Blumer et al., 2003; Webb et al., 2005). Considering that the induction of the AGS proteins is generally cell-type specific, the pathologically defective or actively recovering renal cells may be more permissive for uptake of small molecular reagents (e.g., peptide or nucleic acid sequences) or chemical molecules to manipulate AGS function.

Modulating the protein function of intracellular versus cell-surface receptors in the kidney is obviously more challenging for a variety of factors, largely because of the complex architecture of the kidney with its numerous cell types, irregular blood blow distribution, solute gradients, and wide pH ranges. Each of these factors can complicate the development of an effective therapeutic to modulate AGS. However, the molecular and cellular technologies continue to evolve, leading to new discoveries focusing on effective drug delivery systems. Small molecules are being identified to target specific proteins (Jonasch et al., 2012) or cell types (Raj et al., 2011), which may be a viable approach to develop therapeutics targeting group IV AGS proteins to combat renal cell carcinoma. Alternatively, there are emerging new viral vector systems, such as replication-defective lentiviral vectors (Kim et al., 2010; Park et al., 2010) or adeno-associated virus type 9 (Bostick et al., 2007), which have shown promise for renal cell transduction. These vector systems could be designed to either overexpress small sequences to block Gαi binding or inhibit Gβγ activity, or possibly knockdown the expression levels of AGS genes to control their effect on G protein signaling. Although there has yet to be a successful method to correct renal diseases using these approaches to date, the continued study into the biology of AGS proteins will hopefully elucidate pathways that are crucial to G protein regulation during various renal pathologies.

In summary, the field of AGS protein biology is poised to make significant advances in the kidney over the next few years. With the inevitable convergence between the fields of medicinal chemistry, gene therapy, and G protein biology, new therapies targeting AGS proteins should be considered as potential clinical interventions for acute and chronic kidney diseases.

Abbreviations

AGS

activators of G protein signaling

GAP

GTPase-activating protein

GDI

guanine nucleotide dissociation inhibitor

GEF

guanine nucleotide exchange factor

GPCR

G protein–coupled receptor

GPR

G protein regulatory

RGS

regulators of G protein signaling

Ric-8

resistance to inhibitors of cholinesterase 8

RT-PCR

reverse-transcription polymerase chain reaction

Authorship Contributions

Wrote or contributed to the writing of the manuscript: Park.

Footnotes

This research was supported by the National Institutes of Health National Institute of Diabetes and Digestive and Kidney Diseases [Grant R01-DK09123] and institutional funds.

References

  1. Aksu U, Demirci C, Ince C. (2011) The pathogenesis of acute kidney injury and the toxic triangle of oxygen, reactive oxygen species and nitric oxide. Contrib Nephrol 174:119–128. [DOI] [PubMed] [Google Scholar]
  2. Alexander A, Walker CL. (2011) The role of LKB1 and AMPK in cellular responses to stress and damage. FEBS Lett 585:952–957. [DOI] [PubMed] [Google Scholar]
  3. Anai M, Shojima N, Katagiri H, Ogihara T, Sakoda H, Onishi Y, Ono H, Fujishiro M, Fukushima Y, Horike N, et al. (2005) A novel protein kinase B (PKB)/AKT-binding protein enhances PKB kinase activity and regulates DNA synthesis. J Biol Chem 280:18525–18535. [DOI] [PubMed] [Google Scholar]
  4. Anantharam A, Diversé-Pierluissi MA. (2002) Biochemical approaches to study interaction of calcium channels with RGS12 in primary neuronal cultures. Methods Enzymol 345:60–70. [DOI] [PubMed] [Google Scholar]
  5. Armah HB, Parwani AV. (2010) Xp11.2 translocation renal cell carcinoma. Arch Pathol Lab Med 134:124–129. [DOI] [PubMed] [Google Scholar]
  6. Asano T, Morishita R, Ueda H, Asano M, Kato K. (1998) GTP-binding protein γ12 subunit phosphorylation by protein kinase C–identification of the phosphorylation site and factors involved in cultured cells and rat tissues in vivo. Eur J Biochem 251:314–319. [DOI] [PubMed] [Google Scholar]
  7. Banerjee R, Russo N, Liu M, Basrur V, Bellile E, Palanisamy N, Scanlon CS, van Tubergen E, Inglehart RC, Metwally T, et al. (2014) TRIP13 promotes error-prone nonhomologous end joining and induces chemoresistance in head and neck cancer. Nat Commun 5:4527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Berman DM, Wilkie TM, Gilman AG. (1996) GAIP and RGS4 are GTPase-activating proteins for the Gi subfamily of G protein α subunits. Cell 86:445–452. [DOI] [PubMed] [Google Scholar]
  9. Bernard ML, Peterson YK, Chung P, Jourdan J, Lanier SM. (2001) Selective interaction of AGS3 with G-proteins and the influence of AGS3 on the activation state of G-proteins. J Biol Chem 276:1585–1593. [DOI] [PubMed] [Google Scholar]
  10. Bertolotto C, Lesueur F, Giuliano S, Strub T, de Lichy M, Bille K, Dessen P, d’Hayer B, Mohamdi H, Remenieras A, et al. French Familial Melanoma Study Group (2011) A SUMOylation-defective MITF germline mutation predisposes to melanoma and renal carcinoma. Nature 480:94–98. [DOI] [PubMed] [Google Scholar]
  11. Bertorello A, Aperia A. (1990) Inhibition of proximal tubule Na+-K+-ATPase activity requires simultaneous activation of DA1 and DA2 receptors. Am J Physiol 259:F924–F928. [DOI] [PubMed] [Google Scholar]
  12. Blumer JB, Bernard ML, Peterson YK, Nezu J, Chung P, Dunican DJ, Knoblich JA, Lanier SM. (2003) Interaction of activator of G-protein signaling 3 (AGS3) with LKB1, a serine/threonine kinase involved in cell polarity and cell cycle progression: phosphorylation of the G-protein regulatory (GPR) motif as a regulatory mechanism for the interaction of GPR motifs with Gi α. J Biol Chem 278:23217–23220. [DOI] [PubMed] [Google Scholar]
  13. Blumer JB, Chandler LJ, Lanier SM. (2002) Expression analysis and subcellular distribution of the two G-protein regulators AGS3 and LGN indicate distinct functionality. Localization of LGN to the midbody during cytokinesis. J Biol Chem 277:15897–15903. [DOI] [PubMed] [Google Scholar]
  14. Blumer JB, Lanier SM. (2014) Activators of G protein signaling exhibit broad functionality and define a distinct core signaling triad. Mol Pharmacol 85:388–396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Blumer JB, Smrcka AV, Lanier SM. (2007) Mechanistic pathways and biological roles for receptor-independent activators of G-protein signaling. Pharmacol Ther 113:488–506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Bolcun-Filas E, Rinaldi VD, White ME, Schimenti JC. (2014) Reversal of female infertility by Chk2 ablation reveals the oocyte DNA damage checkpoint pathway. Science 343:533–536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Bonventre JV, Yang L. (2011) Cellular pathophysiology of ischemic acute kidney injury. J Clin Invest 121:4210–4221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Bostick B, Ghosh A, Yue Y, Long C, Duan D. (2007) Systemic AAV-9 transduction in mice is influenced by animal age but not by the route of administration. Gene Ther 14:1605–1609. [DOI] [PubMed] [Google Scholar]
  19. Boucher I, Yu W, Beaudry S, Negoro H, Tran M, Pollak MR, Henderson JM, Denker BM. (2012) Gα12 activation in podocytes leads to cumulative changes in glomerular collagen expression, proteinuria and glomerulosclerosis. Lab Invest 92:662–675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Brann MR, Collins RM, Spiegel A. (1987) Localization of mRNAs encoding the α-subunits of signal-transducing G-proteins within rat brain and among peripheral tissues. FEBS Lett 222:191–198. [DOI] [PubMed] [Google Scholar]
  21. Brogan MD, Behrend EN, Kemppainen RJ. (2001) Regulation of Dexras1 expression by endogenous steroids. Neuroendocrinology 74:244–250. [DOI] [PubMed] [Google Scholar]
  22. Brogi S, Tafi A, Desaubry L, Nebigil CG. (2014) Discovery of GPCR ligands for probing signal transduction pathways. Front Pharmacol 5:255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Brunskill N, Bastani B, Hayes C, Morrissey J, Klahr S. (1991) Localization and polar distribution of several G-protein subunits along nephron segments. Kidney Int 40:997–1006. [DOI] [PubMed] [Google Scholar]
  24. Buscà R, Berra E, Gaggioli C, Khaled M, Bille K, Marchetti B, Thyss R, Fitsialos G, Larribère L, Bertolotto C, et al. (2005) Hypoxia-inducible factor 1α is a new target of microphthalmia-associated transcription factor (MITF) in melanoma cells. J Cell Biol 170:49–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Cali JJ, Balcueva EA, Rybalkin I, Robishaw JD. (1992) Selective tissue distribution of G protein γ subunits, including a new form of the γ subunits identified by cDNA cloning. J Biol Chem 267:24023–24027. [PubMed] [Google Scholar]
  26. Cao X, Cismowski MJ, Sato M, Blumer JB, Lanier SM. (2004) Identification and characterization of AGS4: a protein containing three G-protein regulatory motifs that regulate the activation state of Giα. J Biol Chem 279:27567–27574. [DOI] [PubMed] [Google Scholar]
  27. Chan P, Gabay M, Wright FA, Tall GG. (2011) Ric-8B is a GTP-dependent G protein αs guanine nucleotide exchange factor. J Biol Chem 286:19932–19942. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Chauhan S, Jelen F, Sharina I, Martin E. (2012) The G-protein regulator LGN modulates the activity of the NO receptor soluble guanylate cyclase. Biochem J 446:445–453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Chen J, Futami K, Petillo D, Peng J, Wang P, Knol J, Li Y, Khoo SK, Huang D, Qian CN, et al. (2008) Deficiency of FLCN in mouse kidney led to development of polycystic kidneys and renal neoplasia. PLoS ONE 3:e3581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Ciechanover A. (1993) The ubiquitin-mediated proteolytic pathway. Brain Pathol 3:67–75. [DOI] [PubMed] [Google Scholar]
  31. Cismowski MJ, Lanier SM. (2005) Activation of heterotrimeric G-proteins independent of a G-protein coupled receptor and the implications for signal processing. Rev Physiol Biochem Pharmacol 155:57–80. [DOI] [PubMed] [Google Scholar]
  32. Cismowski MJ, Ma C, Ribas C, Xie X, Spruyt M, Lizano JS, Lanier SM, Duzic E. (2000) Activation of heterotrimeric G-protein signaling by a ras-related protein. Implications for signal integration. J Biol Chem 275:23421–23424. [DOI] [PubMed] [Google Scholar]
  33. Cismowski MJ, Takesono A, Bernard ML, Duzic E, Lanier SM. (2001) Receptor-independent activators of heterotrimeric G-proteins. Life Sci 68:2301–2308. [DOI] [PubMed] [Google Scholar]
  34. Cismowski MJ, Takesono A, Ma C, Lizano JS, Xie X, Fuernkranz H, Lanier SM, Duzic E. (1999) Genetic screens in yeast to identify mammalian nonreceptor modulators of G-protein signaling. Nat Biotechnol 17:878–883. [DOI] [PubMed] [Google Scholar]
  35. Culurgioni S, Alfieri A, Pendolino V, Laddomada F, Mapelli M. (2011) Inscuteable and NuMA proteins bind competitively to Leu-Gly-Asn repeat-enriched protein (LGN) during asymmetric cell divisions. Proc Natl Acad Sci USA 108:20998–21003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Davis IJ, Kim JJ, Ozsolak F, Widlund HR, Rozenblatt-Rosen O, Granter SR, Du J, Fletcher JA, Denny CT, Lessnick SL, et al. (2006) Oncogenic MITF dysregulation in clear cell sarcoma: defining the MiT family of human cancers. Cancer Cell 9:473–484. [DOI] [PubMed] [Google Scholar]
  37. De Vries L, Fischer T, Tronchère H, Brothers GM, Strockbine B, Siderovski DP, Farquhar MG. (2000a) Activator of G protein signaling 3 is a guanine dissociation inhibitor for Gα i subunits. Proc Natl Acad Sci USA 97:14364–14369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. De Vries L, Mousli M, Wurmser A, Farquhar MG. (1995) GAIP, a protein that specifically interacts with the trimeric G protein G α i3, is a member of a protein family with a highly conserved core domain. Proc Natl Acad Sci USA 92:11916–11920. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. De Vries L, Zheng B, Fischer T, Elenko E, Farquhar MG. (2000b) The regulator of G protein signaling family. Annu Rev Pharmacol Toxicol 40:235–271. [DOI] [PubMed] [Google Scholar]
  40. Devarajan P. (2006) Update on mechanisms of ischemic acute kidney injury. J Am Soc Nephrol 17:1503–1520. [DOI] [PubMed] [Google Scholar]
  41. Deveraux Q, Jensen C, Rechsteiner M. (1995) Molecular cloning and expression of a 26 S protease subunit enriched in dileucine repeats. J Biol Chem 270:23726–23729. [DOI] [PubMed] [Google Scholar]
  42. DiBella LM, Benashski SE, Tedford HW, Harrison A, Patel-King RS, King SM. (2001) The Tctex1/Tctex2 class of dynein light chains. Dimerization, differential expression, and interaction with the LC8 protein family. J Biol Chem 276:14366–14373. [DOI] [PubMed] [Google Scholar]
  43. Druey KM, Blumer KJ, Kang VH, Kehrl JH. (1996) Inhibition of G-protein-mediated MAP kinase activation by a new mammalian gene family. Nature 379:742–746. [DOI] [PubMed] [Google Scholar]
  44. El-Mansoury AM, Morgan NG. (1998) Activation of protein kinase C modulates α2-adrenergic signalling in rat pancreatic islets. Cell Signal 10:637–643. [DOI] [PubMed] [Google Scholar]
  45. Falk JD, Vargiu P, Foye PE, Usui H, Perez J, Danielson PE, Lerner DL, Bernal J, Sutcliffe JG. (1999) Rhes: A striatal-specific Ras homolog related to Dexras1. J Neurosci Res 57:782–788. [PubMed] [Google Scholar]
  46. Fan P, Jiang Z, Diamond I, Yao L. (2009) Up-regulation of AGS3 during morphine withdrawal promotes cAMP superactivation via adenylyl cyclase 5 and 7 in rat nucleus accumbens/striatal neurons. Mol Pharmacol 76:526–533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Fang M, Jaffrey SR, Sawa A, Ye K, Luo X, Snyder SH. (2000) Dexras1: a G protein specifically coupled to neuronal nitric oxide synthase via CAPON. Neuron 28:183–193. [DOI] [PubMed] [Google Scholar]
  48. Ferrell K, Deveraux Q, van Nocker S, Rechsteiner M. (1996) Molecular cloning and expression of a multiubiquitin chain binding subunit of the human 26S protease. FEBS Lett 381:143–148. [DOI] [PubMed] [Google Scholar]
  49. Fong HK, Amatruda TT, Birren BW, Simon MI. (1987) Distinct forms of the β subunit of GTP-binding regulatory proteins identified by molecular cloning. Proc Natl Acad Sci USA 84:3792–3796. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Fuja TJ, Schwartz PH, Darcy D, Bryant PJ. (2004) Asymmetric localization of LGN but not AGS3, two homologs of Drosophila pins, in dividing human neural progenitor cells. J Neurosci Res 75:782–793. [DOI] [PubMed] [Google Scholar]
  51. Gabay M, Pinter ME, Wright FA, Chan P, Murphy AJ, Valenzuela DM, Yancopoulos GD, Tall GG. (2011) Ric-8 proteins are molecular chaperones that direct nascent G protein α subunit membrane association. Sci Signal 4:ra79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Gao M, Craig D, Lequin O, Campbell ID, Vogel V, Schulten K. (2003) Structure and functional significance of mechanically unfolded fibronectin type III1 intermediates. Proc Natl Acad Sci USA 100:14784–14789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Garcia-Marcos M, Ear J, Farquhar MG, Ghosh P. (2011) A GDI (AGS3) and a GEF (GIV) regulate autophagy by balancing G protein activity and growth factor signals. Mol Biol Cell 22:673–686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Ghosh P, Beas AO, Bornheimer SJ, Garcia-Marcos M, Forry EP, Johannson C, Ear J, Jung BH, Cabrera B, Carethers JM, et al. (2010) A Gαi-GIV molecular complex binds epidermal growth factor receptor and determines whether cells migrate or proliferate. Mol Biol Cell 21:2338–2354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Ghosh P, Garcia-Marcos M, Bornheimer SJ, Farquhar MG. (2008) Activation of Gαi3 triggers cell migration via regulation of GIV. J Cell Biol 182:381–393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Granter SR, Weilbaecher KN, Quigley C, Fisher DE. (2002) Role for microphthalmia transcription factor in the diagnosis of metastatic malignant melanoma. Appl Immunohistochem Mol Morphol 10:47–51. [DOI] [PubMed] [Google Scholar]
  57. Groves B, Abrahamsen H, Clingan H, Frantz M, Mavor L, Bailey J, Ma D. (2010) An inhibitory role of the G-protein regulator AGS3 in mTOR-dependent macroautophagy. PLoS ONE 5:e8877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Guo R, Liu L, Barajas L. (1998) RT-PCR study of the distribution of connexin 43 mRNA in the glomerulus and renal tubular segments. Am J Physiol 275:R439–R447. [DOI] [PubMed] [Google Scholar]
  59. Gutowski S, Smrcka A, Nowak L, Wu DG, Simon M, Sternweis PC. (1991) Antibodies to the α q subfamily of guanine nucleotide-binding regulatory protein α subunits attenuate activation of phosphatidylinositol 4,5-bisphosphate hydrolysis by hormones. J Biol Chem 266:20519–20524. [PubMed] [Google Scholar]
  60. Haase VH. (2006) Hypoxia-inducible factors in the kidney. Am J Physiol Renal Physiol 291:F271–F281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Hamazaki J, Sasaki K, Kawahara H, Hisanaga S, Tanaka K, Murata S. (2007) Rpn10-mediated degradation of ubiquitinated proteins is essential for mouse development. Mol Cell Biol 27:6629–6638. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Hansen PB, Castrop H, Briggs J, Schnermann J. (2003) Adenosine induces vasoconstriction through Gi-dependent activation of phospholipase C in isolated perfused afferent arterioles of mice. J Am Soc Nephrol 14:2457–2465. [DOI] [PubMed] [Google Scholar]
  63. Harrison LM, He Y. (2011) Rhes and AGS1/Dexras1 affect signaling by dopamine D1 receptors through adenylyl cyclase. J Neurosci Res 89:874–882. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Hong SB, Oh H, Valera VA, Baba M, Schmidt LS, Linehan WM. (2010) Inactivation of the FLCN tumor suppressor gene induces TFE3 transcriptional activity by increasing its nuclear localization. PLoS ONE 5:e15793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Horita S, Seki G, Yamada H, Suzuki M, Koike K, Fujita T. (2013) Roles of renal proximal tubule transport in the pathogenesis of hypertension. Curr Hypertens Rep 9:148–155. [DOI] [PubMed] [Google Scholar]
  66. Hunt TW, Fields TA, Casey PJ, Peralta EG. (1996) RGS10 is a selective activator of G α i GTPase activity. Nature 383:175–177. [DOI] [PubMed] [Google Scholar]
  67. Jonasch E, Futreal PA, Davis IJ, Bailey ST, Kim WY, Brugarolas J, Giaccia AJ, Kurban G, Pause A, Frydman J, et al. (2012) State of the science: an update on renal cell carcinoma. Mol Cancer Res 10:859–880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Kemppainen RJ, Behrend EN. (1998) Dexamethasone rapidly induces a novel ras superfamily member-related gene in AtT-20 cells. J Biol Chem 273:3129–3131. [DOI] [PubMed] [Google Scholar]
  69. Kemppainen RJ, Cox E, Behrend EN, Brogan MD, Ammons JM. (2003) Identification of a glucocorticoid response element in the 3′-flanking region of the human Dexras1 gene. Biochim Biophys Acta 1627:85–89. [DOI] [PubMed] [Google Scholar]
  70. Kim M, Park SW, Kim M, Chen SW, Gerthoffer WT, D’Agati VD, Lee HT. (2010) Selective renal overexpression of human heat shock protein 27 reduces renal ischemia-reperfusion injury in mice. Am J Physiol Renal Physiol 299:F347–F358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Kim WJ, Gersey Z, Daaka Y. (2012) Rap1GAP regulates renal cell carcinoma invasion. Cancer Lett 320:65–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Kimple RJ, De Vries L, Tronchère H, Behe CI, Morris RA, Gist Farquhar M, Siderovski DP. (2001) RGS12 and RGS14 GoLoco motifs are G α(i interaction sites with guanine nucleotide dissociation inhibitor activity. J Biol Chem 276:29275–29281. [DOI] [PubMed] [Google Scholar]
  73. King SM, Barbarese E, Dillman JF, 3rd, Benashski SE, Do KT, Patel-King RS, Pfister KK. (1998) Cytoplasmic dynein contains a family of differentially expressed light chains. Biochemistry 37:15033–15041. [DOI] [PubMed] [Google Scholar]
  74. Koelle MR, Horvitz HR. (1996) EGL-10 regulates G protein signaling in the C. elegans nervous system and shares a conserved domain with many mammalian proteins. Cell 84:115–125. [DOI] [PubMed] [Google Scholar]
  75. Kuiper RP, Schepens M, Thijssen J, Schoenmakers EF, van Kessel AG. (2004) Regulation of the MiTF/TFE bHLH-LZ transcription factors through restricted spatial expression and alternative splicing of functional domains. Nucleic Acids Res 32:2315–2322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Kuiper RP, Schepens M, Thijssen J, van Asseldonk M, van den Berg E, Bridge J, Schuuring E, Schoenmakers EF, van Kessel AG. (2003) Upregulation of the transcription factor TFEB in t(6;11)(p21;q13)-positive renal cell carcinomas due to promoter substitution. Hum Mol Genet 12:1661–1669. [DOI] [PubMed] [Google Scholar]
  77. Kwon M, Pavlov TS, Nozu K, Rasmussen SA, Ilatovskaya DV, Lerch-Gaggl A, North LM, Kim H, Qian F, Sweeney WE, Jr, et al. (2012) G-protein signaling modulator 1 deficiency accelerates cystic disease in an orthologous mouse model of autosomal dominant polycystic kidney disease. Proc Natl Acad Sci USA 109:21462–21467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Lechler T, Fuchs E. (2005) Asymmetric cell divisions promote stratification and differentiation of mammalian skin. Nature 437:275–280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Lee JW, Choi HS, Gyuris J, Brent R, Moore DD. (1995) Two classes of proteins dependent on either the presence or absence of thyroid hormone for interaction with the thyroid hormone receptor. Mol Endocrinol 9:243–254. [DOI] [PubMed] [Google Scholar]
  80. Lenarcyzk M, Regner KR, Park F. (2014a) Localization and expression profile of activators of G-protein signaling in the kidney. FASEB J 28:690.6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Lenarczyk M, Pressly JD, Arnett J, Regner KR, Park F. (2014b) Localization and expression profile of Group I and II AGS proteins in the kidney. J Mol Histol DOI: 10.1007/s10735-014-9605-0. [published ahead of print]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Li A, Saito M, Chuang JZ, Tseng YY, Dedesma C, Tomizawa K, Kaitsuka T, Sung CH. (2011a) Ciliary transition zone activation of phosphorylated Tctex-1 controls ciliary resorption, S-phase entry and fate of neural progenitors. Nat Cell Biol 13:402–411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Li H, Weatherford ET, Davis DR, Keen HL, Grobe JL, Daugherty A, Cassis LA, Allen AM, Sigmund CD. (2011b) Renal proximal tubule angiotensin AT1A receptors regulate blood pressure. Am J Physiol Regul Integr Comp Physiol 301:R1067–R1077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Li XC, Schimenti JC. (2007) Mouse pachytene checkpoint 2 (trip13) is required for completing meiotic recombination but not synapsis. PLoS Genet 3:e130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Lin Y, Smrcka AV. (2011) Understanding molecular recognition by G protein βγ subunits on the path to pharmacological targeting. Mol Pharmacol 80:551–557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Liu CT, Garnaas MK, Tin A, Kottgen A, Franceschini N, Peralta CA, de Boer IH, Lu X, Atkinson E, Ding J, et al. CKDGen Consortium (2011) Genetic association for renal traits among participants of African ancestry reveals new loci for renal function. PLoS Genet 7:e1002264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Maldonado-Agurto R, Toro G, Fuentealba J, Arriagada C, Campos T, Albistur M, Henriquez JP, Olate J, Hinrichs MV, Torrejón M. (2011) Cloning and spatiotemporal expression of RIC-8 in Xenopus embryogenesis. Gene Expr Patterns 11:401–408. [DOI] [PubMed] [Google Scholar]
  88. Mathur M, Samuels HH. (2007) Role of PSF-TFE3 oncoprotein in the development of papillary renal cell carcinomas. Oncogene 26:277–283. [DOI] [PubMed] [Google Scholar]
  89. Meloni AM, Dobbs RM, Pontes JE, Sandberg AA. (1993) Translocation (X;1) in papillary renal cell carcinoma. A new cytogenetic subtype. Cancer Genet Cytogenet 65:1–6. [DOI] [PubMed] [Google Scholar]
  90. Miller KG, Emerson MD, McManus JR, Rand JB. (2000) RIC-8 (Synembryn): a novel conserved protein that is required for G(q)α signaling in the C. elegans nervous system. Neuron 27:289–299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Mittal V, Linder ME. (2004) The RGS14 GoLoco domain discriminates among Gαi isoforms. J Biol Chem 279:46772–46778. [DOI] [PubMed] [Google Scholar]
  92. Miyamoto KI, Itho M. (2001) Transcriptional regulation of the NPT2 gene by dietary phosphate. Kidney Int 60:412–415. [DOI] [PubMed] [Google Scholar]
  93. Nadella R, Blumer JB, Jia G, Kwon M, Akbulut T, Qian F, Sedlic F, Wakatsuki T, Sweeney WE, Jr, Wilson PD, et al. (2010) Activator of G protein signaling 3 promotes epithelial cell proliferation in PKD. J Am Soc Nephrol 21:1275–1280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Neer EJ, Lok JM, Wolf LG. (1984) Purification and properties of the inhibitory guanine nucleotide regulatory unit of brain adenylate cyclase. J Biol Chem 259:14222–14229. [PubMed] [Google Scholar]
  95. Nguyen CH, Watts VJ. (2006) Dexamethasone-induced Ras protein 1 negatively regulates protein kinase C δ: implications for adenylyl cyclase 2 signaling. Mol Pharmacol 69:1763–1771. [DOI] [PubMed] [Google Scholar]
  96. Nielsen S, DiGiovanni SR, Christensen EI, Knepper MA, Harris HW. (1993) Cellular and subcellular immunolocalization of vasopressin-regulated water channel in rat kidney. Proc Natl Acad Sci USA 90:11663–11667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Nitta K, Uchida K, Kawashima A, Tsutsui T, Ozu H, Naito T, Yumura W, Nihei H. (1994) Identification of GTP-binding proteins in human glomeruli. Nippon Jinzo Gakkai Shi 36:9–12. [PubMed] [Google Scholar]
  98. Nordquist DT, Kozak CA, Orr HT. (1988) cDNA cloning and characterization of three genes uniquely expressed in cerebellum by Purkinje neurons. J Neurosci 8:4780-4789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. O’Connor PM, Cowley AW., Jr (2010) Modulation of pressure-natriuresis by renal medullary reactive oxygen species and nitric oxide. Curr Hypertens Rep 12:86–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Oner SS, An N, Vural A, Breton B, Bouvier M, Blumer JB, Lanier SM. (2010) Regulation of the AGS3·Gαi signaling complex by a seven-transmembrane span receptor. J Biol Chem 285:33949–33958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Oner SS, Vural A, Lanier SM. (2013) Translocation of activator of G-protein signaling 3 to the Golgi apparatus in response to receptor activation and its effect on the trans-Golgi network. J Biol Chem 288:24091–24103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Park SW, Chen SW, Kim M, D'Agati VD, Lee HT. (2010) Selective intrarenal human A1 adenosine receptor overexpression reduces acute liver and kidney injury after hepatic ischemia reperfusion in mice. Lab Invest 90:476–495. [DOI] [PubMed] [Google Scholar]
  103. Pattingre S, De Vries L, Bauvy C, Chantret I, Cluzeaud F, Ogier-Denis E, Vandewalle A, Codogno P. (2003) The G-protein regulator AGS3 controls an early event during macroautophagy in human intestinal HT-29 cells. J Biol Chem 278:20995–21002. [DOI] [PubMed] [Google Scholar]
  104. Pattingre S, Petiot A, Codogno P. (2004) Analyses of Gα-interacting protein and activator of G-protein-signaling-3 functions in macroautophagy. Methods Enzymol 390:17–31. [DOI] [PubMed] [Google Scholar]
  105. Peterson YK, Bernard ML, Ma H, Hazard S, 3rd, Graber SG, Lanier SM. (2000) Stabilization of the GDP-bound conformation of Giα by a peptide derived from the G-protein regulatory motif of AGS3. J Biol Chem 275:33193–33196. [DOI] [PubMed] [Google Scholar]
  106. Pippin JW, Brinkkoetter PT, Cormack-Aboud FC, Durvasula RV, Hauser PV, Kowalewska J, Krofft RD, Logar CM, Marshall CB, Ohse T, et al. (2009) Inducible rodent models of acquired podocyte diseases. Am J Physiol Renal Physiol 296:F213–F229. [DOI] [PubMed] [Google Scholar]
  107. Pizzinat N, Takesono A, Lanier SM. (2001) Identification of a truncated form of the G-protein regulator AGS3 in heart that lacks the tetratricopeptide repeat domains. J Biol Chem 276:16601–16610. [DOI] [PubMed] [Google Scholar]
  108. Potla U, Ni J, Vadaparampil J, Yang G, Leventhal JS, Campbell KN, Chuang PY, Morozov A, He JC, D’Agati VD, et al. (2014) Podocyte-specific RAP1GAP expression contributes to focal segmental glomerulosclerosis-associated glomerular injury. J Clin Invest 124:1757–1769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Pusch W, Jähner D, Ivell R. (1998) Molecular cloning and testicular expression of the gene transcripts encoding the murine multiubiquitin-chain-binding protein (Mcb1). Gene 207:19–24. [DOI] [PubMed] [Google Scholar]
  110. Raj L, Ide T, Gurkar AU, Foley M, Schenone M, Li X, Tolliday NJ, Golub TR, Carr SA, Shamji AF, et al. (2011) Selective killing of cancer cells by a small molecule targeting the stress response to ROS. Nature 475:231–234. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  111. Rao Q, Liu B, Cheng L, Zhu Y, Shi QL, Wu B, Jiang SJ, Wang Y, Wang X, Yu B, et al. (2012) Renal cell carcinomas with t(6;11)(p21;q12): A clinicopathologic study emphasizing unusual morphology, novel α-TFEB gene fusion point, immunobiomarkers, and ultrastructural features, as well as detection of the gene fusion by fluorescence in situ hybridization. Am J Surg Pathol 36:1327–1338. [DOI] [PubMed] [Google Scholar]
  112. Ray K, Kunsch C, Bonner LM, Robishaw JD. (1995) Isolation of cDNA clones encoding eight different human G protein γ subunits, including three novel forms designated the γ 4, γ 10, and γ 11 subunits. J Biol Chem 270:21765–21771. [DOI] [PubMed] [Google Scholar]
  113. Regner KR, Nozu K, Lanier SM, Blumer JB, Avner ED, Sweeney WE, Jr, Park F. (2011) Loss of activator of G-protein signaling 3 impairs renal tubular regeneration following acute kidney injury in rodents. FASEB J 25:1844–1855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Reif K, Cyster JG. (2000) RGS molecule expression in murine B lymphocytes and ability to down-regulate chemotaxis to lymphoid chemokines. J Immunol 164:4720–4729. [DOI] [PubMed] [Google Scholar]
  115. Richman RW, Diversé-Pierluissi MA. (2004) Mapping of RGS12-Cav2.2 channel interaction. Methods Enzymol 390:224–239. [DOI] [PubMed] [Google Scholar]
  116. Richman RW, Strock J, Hains MD, Cabanilla NJ, Lau KK, Siderovski DP, Diversé-Pierluissi M. (2005) RGS12 interacts with the SNARE-binding region of the Cav2.2 calcium channel. J Biol Chem 280:1521–1528. [DOI] [PubMed] [Google Scholar]
  117. Roig I, Dowdle JA, Toth A, de Rooij DG, Jasin M, Keeney S. (2010) Mouse TRIP13/PCH2 is required for recombination and normal higher-order chromosome structure during meiosis. PLoS Genet 6:e1001062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Ruan X, Chatziantoniou C, Arendshorst WJ. (1999) Impaired prostaglandin E2/prostaglandin I2 receptor-Gs protein interactions in isolated renal resistance arterioles of spontaneously hypertensive rats. Hypertension 34:1134–1140. [DOI] [PubMed] [Google Scholar]
  119. Rubinfeld B, Munemitsu S, Clark R, Conroy L, Watt K, Crosier WJ, McCormick F, Polakis P. (1991) Molecular cloning of a GTPase activating protein specific for the Krev-1 protein p21rap1. Cell 65:1033–1042. [DOI] [PubMed] [Google Scholar]
  120. Rubio JP, Levy ER, Dobson-Stone C, Monaco AP. (1999) Genomic organization of the human gα14 and Gαq genes and mutation analysis in chorea-acanthocytosis (CHAC). Genomics 57:84–93. [DOI] [PubMed] [Google Scholar]
  121. Rusai K, Prokai A, Juanxing C, Meszaros K, Szalay B, Pásti K, Müller V, Heemann U, Lutz J, Tulassay T, et al. (2013) Dexamethasone protects from renal ischemia/reperfusion injury: a possible association with SGK-1. Acta Physiol Hung 100:173–185. [DOI] [PubMed] [Google Scholar]
  122. Saito H, Tsumura H, Otake S, Nishida A, Furukawa T, Suzuki N. (2005) L7/Pcp-2-specific expression of Cre recombinase using knock-in approach. Biochem Biophys Res Commun 331:1216–1221. [DOI] [PubMed] [Google Scholar]
  123. Sanada K, Tsai LH. (2005) G protein βγ subunits and AGS3 control spindle orientation and asymmetric cell fate of cerebral cortical progenitors. Cell 122:119–131. [DOI] [PubMed] [Google Scholar]
  124. Sano T, Ohyama K, Yamano Y, Nakagomi Y, Nakazawa S, Kikyo M, Shirai H, Blank JS, Exton JH, Inagami T. (1997) A domain for G protein coupling in carboxyl-terminal tail of rat angiotensin II receptor type 1A. J Biol Chem 272:23631–23636. [DOI] [PubMed] [Google Scholar]
  125. Sato M, Blumer JB, Simon V, Lanier SM. (2006a) Accessory proteins for G proteins: partners in signaling. Annu Rev Pharmacol Toxicol 46:151–187. [DOI] [PubMed] [Google Scholar]
  126. Sato M, Cismowski MJ, Toyota E, Smrcka AV, Lucchesi PA, Chilian WM, Lanier SM. (2006b) Identification of a receptor-independent activator of G protein signaling (AGS8) in ischemic heart and its interaction with Gβγ. Proc Natl Acad Sci USA 103:797–802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Sato M, Gettys TW, Lanier SM. (2004) AGS3 and signal integration by Gαs- and Gαi-coupled receptors: AGS3 blocks the sensitization of adenylyl cyclase following prolonged stimulation of a Gαi-coupled receptor by influencing processing of Gαi. J Biol Chem 279:13375–13382. [DOI] [PubMed] [Google Scholar]
  128. Sato M, Hiraoka M, Suzuki H, Bai Y, Kurotani R, Yokoyama U, Okumura S, Cismowski MJ, Lanier SM, Ishikawa Y. (2011) Identification of transcription factor E3 (TFE3) as a receptor-independent activator of Gα16: gene regulation by nuclear Gα subunit and its activator. J Biol Chem 286:17766–17776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Sato M, Hiraoka M, Suzuki H, Sakima M, Mamun AA, Yamane Y, Fujita T, Yokoyama U, Okumura S, Ishikawa Y. (2014) Protection of cardiomyocytes from the hypoxia-mediated injury by a peptide targeting the activator of G-protein signaling 8. PLoS ONE 9:e91980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Sato M, Jiao Q, Honda T, Kurotani R, Toyota E, Okumura S, Takeya T, Minamisawa S, Lanier SM, Ishikawa Y. (2009) Activator of G protein signaling 8 (AGS8) is required for hypoxia-induced apoptosis of cardiomyocytes: role of G βγ and connexin 43 (CX43). J Biol Chem 284:31431–31440. [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Schiff ML, Siderovski DP, Jordan JD, Brothers G, Snow B, De Vries L, Ortiz DF, Diversé-Pierluissi M. (2000) Tyrosine-kinase-dependent recruitment of RGS12 to the N-type calcium channel. Nature 408:723–727. [DOI] [PubMed] [Google Scholar]
  132. Senkfor SI, Johnson GL, Berl T. (1993) A molecular map of G protein α chains in microdissected rat nephron segments. J Clin Invest 92:786–790. [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Siderovski DP, Diversé-Pierluissi Ma, De Vries L. (1999) The GoLoco motif: a Gαi/o binding motif and potential guanine-nucleotide exchange factor. Trends Biochem Sci 24:340–341. [DOI] [PubMed] [Google Scholar]
  134. Siderovski DP, Hessel A, Chung S, Mak TW, Tyers M. (1996) A new family of regulators of G-protein-coupled receptors? Curr Biol 6:211–212. [DOI] [PubMed] [Google Scholar]
  135. Smrcka AV. (2008) G protein βγ subunits: central mediators of G protein-coupled receptor signaling. Cell Mol Life Sci 65:2191–2214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Smrcka AV, Lehmann DM, Dessal AL. (2008) G protein βγ subunits as targets for small molecule therapeutic development. Comb Chem High Throughput Screen 11:382–395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Snow BE, Antonio L, Suggs S, Gutstein HB, Siderovski DP. (1997) Molecular cloning and expression analysis of rat Rgs12 and Rgs14. Biochem Biophys Res Commun 233:770–777. [DOI] [PubMed] [Google Scholar]
  138. Snow BE, Betts L, Mangion J, Sondek J, Siderovski DP. (1999) Fidelity of G protein β-subunit association by the G protein γ-subunit-like domains of RGS6, RGS7, and RGS11. Proc Natl Acad Sci USA 96:6489–6494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Snow BE, Hall RA, Krumins AM, Brothers GM, Bouchard D, Brothers CA, Chung S, Mangion J, Gilman AG, Lefkowitz RJ, et al. (1998) GTPase activating specificity of RGS12 and binding specificity of an alternatively spliced PDZ (PSD-95/Dlg/ZO-1) domain. J Biol Chem 273:17749–17755. [DOI] [PubMed] [Google Scholar]
  140. Spano D, Branchi I, Rosica A, Pirro MT, Riccio A, Mithbaokar P, Affuso A, Arra C, Campolongo P, Terracciano D, et al. (2004) Rhes is involved in striatal function. Mol Cell Biol 24:5788–5796. [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Sternweis PC, Robishaw JD. (1984) Isolation of two proteins with high affinity for guanine nucleotides from membranes of bovine brain. J Biol Chem 259:13806–13813. [PubMed] [Google Scholar]
  142. Stow JL, Sabolic I, Brown D. (1991) Heterogeneous localization of G protein α-subunits in rat kidney. Am J Physiol 261:F831–F840. [DOI] [PubMed] [Google Scholar]
  143. Strathmann M, Wilkie TM, Simon MI. (1989) Diversity of the G-protein family: sequences from five additional α subunits in the mouse. Proc Natl Acad Sci USA 86:7407–7409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Strathmann M, Wilkie TM, Simon MI. (1990) Alternative splicing produces transcripts encoding two forms of the α subunit of GTP-binding protein Go. Proc Natl Acad Sci USA 87:6477–6481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Takesono A, Cismowski MJ, Ribas C, Bernard M, Chung P, Hazard S, 3rd, Duzic E, Lanier SM. (1999) Receptor-independent activators of heterotrimeric G-protein signaling pathways. J Biol Chem 274:33202–33205. [DOI] [PubMed] [Google Scholar]
  146. Tall GG, Krumins AM, Gilman AG. (2003) Mammalian Ric-8A (synembryn) is a heterotrimeric Gα protein guanine nucleotide exchange factor. J Biol Chem 278:8356–8362. [DOI] [PubMed] [Google Scholar]
  147. Tan JJ, Ong SA, Chen KS. (2011) Rasd1 interacts with Ear2 (Nr2f6) to regulate renin transcription. BMC Mol Biol 12:4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Tanaka K. (1995) Molecular biology of proteasomes. Mol Biol Rep 21:21–26. [DOI] [PubMed] [Google Scholar]
  149. Thapliyal A, Bannister RA, Hanks C, Adams BA. (2008) The monomeric G proteins AGS1 and Rhes selectively influence Gαi-dependent signaling to modulate N-type (CaV2.2) calcium channels. Am J Physiol Cell Physiol 295:C1417–C1426. [DOI] [PubMed] [Google Scholar]
  150. Tipton AR, Wang K, Oladimeji P, Sufi S, Gu Z, Liu ST. (2012) Identification of novel mitosis regulators through data mining with human centromere/kinetochore proteins as group queries. BMC Cell Biol 13:15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Toda N, Okamura T. (2011) Modulation of renal blood flow and vascular tone by neuronal nitric oxide synthase-derived nitric oxide. J Vasc Res 48:1–10. [DOI] [PubMed] [Google Scholar]
  152. Tõnissoo T, Kõks S, Meier R, Raud S, Plaas M, Vasar E, Karis A. (2006) Heterozygous mice with Ric-8 mutation exhibit impaired spatial memory and decreased anxiety. Behav Brain Res 167:42–48. [DOI] [PubMed] [Google Scholar]
  153. Tõnissoo T, Meier R, Talts K, Plaas M, Karis A. (2003) Expression of ric-8 (synembryn) gene in the nervous system of developing and adult mouse. Gene Expr Patterns 3:591–594. [DOI] [PubMed] [Google Scholar]
  154. Traver S, Bidot C, Spassky N, Baltauss T, De Tand MF, Thomas JL, Zalc B, Janoueix-Lerosey I, Gunzburg JD. (2000) RGS14 is a novel Rap effector that preferentially regulates the GTPase activity of gαo. Biochem J 350:19–29. [PMC free article] [PubMed] [Google Scholar]
  155. Tu Y, Wu C. (1999) Cloning, expression and characterization of a novel human Ras-related protein that is regulated by glucocorticoid hormone. Biochim Biophys Acta 1489:452–456. [DOI] [PubMed] [Google Scholar]
  156. Tummala H, Fleming S, Hocking PM, Wehner D, Naseem Z, Ali M, Inglehearn CF, Zhelev N, Lester DH. (2011) The D153del mutation in GNB3 gene causes tissue specific signalling patterns and an abnormal renal morphology in Rge chickens. PLoS ONE 6:e21156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  157. Vaidyanathan G, Cismowski MJ, Wang G, Vincent TS, Brown KD, Lanier SM. (2004) The Ras-related protein AGS1/RASD1 suppresses cell growth. Oncogene 23:5858–5863. [DOI] [PubMed] [Google Scholar]
  158. Vargiu P, De Abajo R, Garcia-Ranea JA, Valencia A, Santisteban P, Crespo P, Bernal J. (2004) The small GTP-binding protein, Rhes, regulates signal transduction from G protein-coupled receptors. Oncogene 23:559–568. [DOI] [PubMed] [Google Scholar]
  159. Verma R, Oania R, Graumann J, Deshaies RJ. (2004) Multiubiquitin chain receptors define a layer of substrate selectivity in the ubiquitin-proteasome system. Cell 118:99–110. [DOI] [PubMed] [Google Scholar]
  160. von Weizsäcker E, Strathmann MP, Simon MI. (1992) Diversity among the β subunits of heterotrimeric GTP-binding proteins: characterization of a novel β-subunit cDNA. Biochem Biophys Res Commun 183:350–356. [DOI] [PubMed] [Google Scholar]
  161. Vural A, McQuiston TJ, Blumer JB, Park C, Hwang IY, Williams-Bey Y, Shi CS, Ma DZ, Kehrl JH. (2013) Normal autophagic activity in macrophages from mice lacking Gαi3, AGS3, or RGS19. PLoS ONE 8:e81886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  162. Wang H, Misaki T, Taupin V, Eguchi A, Ghosh P, Farquhar MG. (2015) GIV/Girdin links vascular endothelial growth factor signaling to Akt survival signaling in podocytes independent of nephrin. J Am Soc Nephrol 26:314–327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Wang K, Sturt-Gillespie B, Hittle JC, Macdonald D, Chan GK, Yen TJ, Liu ST. (2014) Thyroid hormone receptor interacting protein 13 (TRIP13) AAA-ATPase is a novel mitotic checkpoint-silencing protein. J Biol Chem 289:23928–23937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. Watson N, Linder ME, Druey KM, Kehrl JH, Blumer KJ. (1996) RGS family members: GTPase-activating proteins for heterotrimeric G-protein α-subunits. Nature 383:172–175. [DOI] [PubMed] [Google Scholar]
  165. Webb CK, McCudden CR, Willard FS, Kimple RJ, Siderovski DP, Oxford GS. (2005) D2 dopamine receptor activation of potassium channels is selectively decoupled by Gα-specific GoLoco motif peptides. J Neurochem 92:1408–1418. [DOI] [PubMed] [Google Scholar]
  166. Weterman MA, van Groningen JJ, den Hartog A, Geurts van Kessel A. (2001) Transformation capacities of the papillary renal cell carcinoma-associated PRCCTFE3 and TFE3PRCC fusion genes. Oncogene 20:1414–1424. [DOI] [PubMed] [Google Scholar]
  167. Weterman MA, Wilbrink M, Geurts van Kessel A. (1996a) Fusion of the transcription factor TFE3 gene to a novel gene, PRCC, in t(X;1)(p11;q21)-positive papillary renal cell carcinomas. Proc Natl Acad Sci USA 93:15294–15298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  168. Weterman MA, Wilbrink M, Janssen I, Janssen HA, van den Berg E, Fisher SE, Craig I, Geurts van Kessel A. (1996b) Molecular cloning of the papillary renal cell carcinoma-associated translocation (X;1)(p11;q21) breakpoint. Cytogenet Cell Genet 75:2–6. [DOI] [PubMed] [Google Scholar]
  169. Weterman MJ, van Groningen JJ, Jansen A, van Kessel AG. (2000) Nuclear localization and transactivating capacities of the papillary renal cell carcinoma-associated TFE3 and PRCC (fusion) proteins. Oncogene 19:69–74. [DOI] [PubMed] [Google Scholar]
  170. Wettschureck N, Offermanns S. (2005) Mammalian G proteins and their cell type specific functions. Physiol Rev 85:1159–1204. [DOI] [PubMed] [Google Scholar]
  171. Wilkie TM, Scherle PA, Strathmann MP, Slepak VZ, Simon MI. (1991) Characterization of G-protein α subunits in the Gq class: expression in murine tissues and in stromal and hematopoietic cell lines. Proc Natl Acad Sci USA 88:10049–10053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  172. Williamson CM, Schofield J, Dutton ER, Seymour A, Beechey CV, Edwards YH, Peters J. (1996) Glomerular-specific imprinting of the mouse gsα gene: how does this relate to hormone resistance in albright hereditary osteodystrophy? Genomics 36:280–287. [DOI] [PubMed] [Google Scholar]
  173. Xiao Z, Wan Q, Du Q, Zheng Z. (2012) Gα/LGN-mediated asymmetric spindle positioning does not lead to unequal cleavage of the mother cell in 3-D cultured MDCK cells. Biochem Biophys Res Commun 420:888–894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  174. Yanagisawa H, Kurihara N, Klahr S, Morrissey J, Wada O. (1994) Regional characterization of G-protein subunits in glomeruli, cortices and medullas of the rat kidney. Nephron 66:447–452. [DOI] [PubMed] [Google Scholar]
  175. Yanagisawa H, Morrissey J, Klahr S. (1993) Bilateral ureteral obstruction alters levels of the G-protein subunits G α s and G α q/11. Kidney Int 43:865–871. [DOI] [PubMed] [Google Scholar]
  176. Yang S, Li YP. (2007) RGS12 is essential for RANKL-evoked signaling for terminal differentiation of osteoclasts in vitro. J Bone Miner Res 22:45–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  177. Yeh C, Li A, Chuang JZ, Saito M, Cáceres A, Sung CH. (2013) IGF-1 activates a cilium-localized noncanonical Gβγ signaling pathway that regulates cell-cycle progression. Dev Cell 26:358–368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  178. Yu AS. (2015) Claudins and the kidney. J Am Soc Nephrol 26:11–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  179. Yu S, Yu D, Lee E, Eckhaus M, Lee R, Corria Z, Accili D, Westphal H, Weinstein LS. (1998) Variable and tissue-specific hormone resistance in heterotrimeric Gs protein α-subunit (Gsα) knockout mice is due to tissue-specific imprinting of the gsα gene. Proc Natl Acad Sci USA 95:8715–8720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  180. Zhang Y, Jose PA, Zeng C. (2011) Regulation of sodium transport in the proximal tubule by endothelin. Contrib Nephrol 172:63–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  181. Zhao P, Nguyen CH, Chidiac P. (2010) The proline-rich N-terminal domain of G18 exhibits a novel G protein regulatory function. J Biol Chem 285:9008–9017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  182. Zheng S, Yu P, Zeng C, Wang Z, Yang Z, Andrews PM, Felder RA, Jose PA. (2003) Gα12- and Gα13-protein subunit linkage of D5 dopamine receptors in the nephron. Hypertension 41:604–610. [DOI] [PubMed] [Google Scholar]
  183. Zheng Z, Zhu H, Wan Q, Liu J, Xiao Z, Siderovski DP, Du Q. (2010) LGN regulates mitotic spindle orientation during epithelial morphogenesis. J Cell Biol 189:275–288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  184. Zhu J, Wen W, Zheng Z, Shang Y, Wei Z, Xiao Z, Pan Z, Du Q, Wang W, Zhang M. (2011) LGN/mInsc and LGN/NuMA complex structures suggest distinct functions in asymmetric cell division for the Par3/mInsc/LGN and Gαi/LGN/NuMA pathways. Mol Cell 43:418–431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  185. Zhuo JL, Li XC. (2011) New insights and perspectives on intrarenal renin-angiotensin system: focus on intracrine/intracellular angiotensin II. Peptides 32:1551–1565. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from The Journal of Pharmacology and Experimental Therapeutics are provided here courtesy of American Society for Pharmacology and Experimental Therapeutics

RESOURCES