Skip to main content
American Journal of Physiology - Renal Physiology logoLink to American Journal of Physiology - Renal Physiology
. 2021 May 24;321(1):F50–F68. doi: 10.1152/ajprenal.00077.2021

Landscape of GPCR expression along the mouse nephron

Brian G Poll 1, Lihe Chen 1, Chung-Lin Chou 1, Viswanathan Raghuram 1, Mark A Knepper 1,
PMCID: PMC8321805  PMID: 34029142

graphic file with name f-00077-2021r01.jpg

Keywords: G protein-coupled receptors, nephron transport, omics

Abstract

Kidney transport and other renal functions are regulated by multiple G protein-coupled receptors (GPCRs) expressed along the renal tubule. The rapid, recent appearance of comprehensive unbiased gene expression data in the various renal tubule segments, chiefly RNA sequencing and protein mass spectrometry data, has provided a means of identifying patterns of GPCR expression along the renal tubule. To allow for comprehensive mapping, we first curated a comprehensive list of GPCRs in the genomes of mice, rats, and humans (https://hpcwebapps.cit.nih.gov/ESBL/Database/GPCRs/) using multiple online data sources. We used this list to mine segment-specific and cell type-specific expression data from RNA-sequencing studies in microdissected mouse tubule segments to identify GPCRs that are selectively expressed in discrete tubule segments. Comparisons of these mapped mouse GPCRs with other omics datasets as well as functional data from isolated perfused tubule and micropuncture studies confirmed patterns of expression for well-known receptors and identified poorly studied GPCRs that are likely to play roles in the regulation of renal tubule function. Thus, we provide data resources for GPCR expression across the renal tubule, highlighting both well-known GPCRs and understudied receptors to provide guidance for future studies.

INTRODUCTION

G protein-coupled receptor (GPCR) signaling plays a crucial role in renal function. Many key GPCRs expressed in renal tubule epithelia were initially identified through biochemical studies in isolated, microdissected renal tubules largely in the laboratory of Morel et al. (1, 2). Among other receptors, these studies localized the parathyroid hormone (PTH) receptor to the proximal tubule (PT) and thick ascending limb (TAL) (3), glucagon (4) and calcitonin receptors (5) to the TAL (6), and the vasopressin receptor to the collecting duct (CD) (1, 7). These and other studies of the same era defined an early pattern of GPCR expression along the renal tubule, geared chiefly to the regulation of transport, as demonstrated in isolated perfused tubule studies (8).

In general, all of these previously characterized receptors were identified through hypothesis-driven studies. Since the publication of various genome projects around 2,000, progress in identifying previously unsuspected gene products has been accelerated by the development of unbiased “-omics” approaches such as mass spectrometry-based proteomics and next-generation sequencing-based transcriptomics. Examples include transcriptomics in microdissected renal tubules (911), single-cell transcriptomics (10, 1215), and proteomics in microdissected tubules (16). These studies have yielded a wealth of omics data, much of which is publicly accessible (see, for example, https://hpcwebapps.cit.nih.gov/ESBL/Database/) and available for hypothesis generation and analysis.

Insel et al. (17, 18) have proposed the use of unbiased comprehensive omics datasets to identify novel patterns of GPCR expression (GPCRomics) as tools for the discovery of new physiological functions that could lead to therapeutic drug targets. Although the work of several laboratories, including our own, has generated a wealth of omics data, these datasets have not been examined through a broad lens, particularly with respect to large gene families such as GPCRs. After creating a curated database of mouse GPCRs, we mapped the expression of 758 GPCRs, plus frizzled receptors, across 14 nephron segments using existing transcriptomic and proteomic data. From these data, we highlighted GPCRs with selective expression patterns in discrete tubule segments. The objective of this review is to integrate omics data and the existing literature to describe the landscape of GPCR expression among the segments of the renal tubule.

MAPPING GPCRs ACROSS THE NEPHRON

To map GPCR expression, we curated a complete list of mouse GPCRs (Fig. 1A) using multiple online databases. The resulting GPCR lists for the mouse, along with their rat and human orthologs, are provided at https://hpcwebapps.cit.nih.gov/ESBL/Database/GPCRs/. This curated GPCR list was mapped to RNA-sequencing (RNA-seq) data from 14 microdissected tubule segments and the glomerulus generated by Chen et al. (9) (Fig. 1, B and C). Expression levels for all these GPCRs are provided at https://hpcwebapps.cit.nih.gov/ESBL/Database/GPCRs/TubuleTPM.html. This yielded matches to 758 GPCRs across the renal tubule. From this extensive list, we ranked GPCR expression in each microdissected segment. The top 10 GPCRs expressed in each segment is shown in Table 1. From these data, we highlighted GPCRs that are selectively expressed in discrete tubule regions (Fig. 1D), defined as GPCR expression in one tubule region being at least 1.5 times greater than any other point along the renal tubule. This threshold was chosen to identify receptors with the potential for having functional roles in particular tubule segments. Several of the most highly expressed GPCRs in the renal tubule (Table 1), such as G protein-coupled receptor class C group 5 member C (Gprc5c) and adhesion G protein-coupled receptor G1 (Adgrg1), were expressed ubiquitously, suggesting nonspecific functions, i.e., housekeeping genes. This review will focus on GPCRs that were detected to be expressed in transcripts per kilobase million (TPM) value of 10 or greater, which corresponds to a rank of 105 of 498 GPCRs detected at ≥1.0 TPM. This criterion highlights the GPCRs most likely to be detected at the protein level. Additional details about how we cataloged and curated the GPCRs we present in this review, along with the cross-referenced datasets, are detailed in the following sections (see appendix).

Figure 1.

Figure 1.

Creating a database of G protein-coupled receptors (GPCRs) and filtering for segment-specific expression. GPCR gene entries were combined from Mouse Genome Informatics (MGI), International Union of Basic and Clinical Pharmacology/British Pharmacological Society Guide to Pharmacology (IUPHAR), and Ensembl to create a complete GPCR database for mouse, rat, and human orthologs (A). This list was then mapped to RNA-sequencing (RNA-seq) data from 14 microdissected renal tubule segments (B) to get a complete picture of GPCR expression (C). From this mapped GPCR expression, segment-specific GPCRs were determined (D). These GPCRs were then compared with existing literature knowledge and cross referenced with other existing omics data sets. IMCD, inner medullary collecting duct; OMCD, outer medullary collecting duct; TPM, transcripts per kilobase million.

Table 1.

Most highly expressed G protein-coupled receptors in each tubule segment

Rank PTS1 PTS2 PTS3 DTL1 DTL2 DTL3 ATL MTAL CTAL DCT CNT CCD OMCD IMCD Glom
1 Pth1r Pth1r Pth1r Adgrg1 Adgrg1 Adgrg1 Adgrg1 Ptger3 Adgrg1 Pth1r Adgrg1 Adgrg1 Adgrg1 Avpr2 Pth1r
370 970 544 1,055 830 1,136 645 729 928 569 1,055 1,304 1,242 1,012 1,089
2 Gprc5c Cckar Gprc5c Gprc5c Ackr3 Gprc5c Avpr2 Adgrg1 Pth1r Adgrg1 Avpr2 Avpr2 Avpr2 Ptger1 F2r
177 259 128 320 460 917 189 600 585 303 631 1,011 1,214 336 463
3 Adgrg1 Gprc5c Tm2d1 Gprc5a Gprc5c Gprc5a Adgre5 Gcgr Ptger3 Ptgfr Gprc5c Gprc5c Gprc5c Adgrg1 Adgre5
97 236 115 315 329 285 120 289 512 213 397 419 562 187 422
4 Lpar3 Tm2d1 Cckar Adgrl4 Gprc5a Adora1 Tm2d1 Avpr2 Gcgr Gprc5c Oxgr1 Avpr1a Ptger1 Gprc5c Agtr1a
94 117 114 261 221 163 111 137 488 208 332 229 495 168 396
5 Tm2d1 Adgrg1 Adrb2 Ednrb Adora1 F2rl1 F2r Gprc5c Casr Oxgr1 Avpr1a Oxgr1 Adgrf5 Tm2d1 Adgrf5
82 63 46 217 190 144 92 130 225 174 301 206 406 162 231
6 Agtr1a Adgra3 Adgrg1 F2rl1 Adgrl4 Tm2d1 Adora1 Tm2d1 Gprc5c Gcgr Adgrf5 Adgrf5 P2ry14 P2ry14 S1pr3
52 58 36 115 174 125 63 127 195 160 155 186 237 146 219
7 Lgr4 Sucnr1 Gpr137 Adgrf5 Tm2d1 Lgr4 Ptger3 Fzd4 Tm2d1 Tm2d1 Tm2d1 Ptgfr Tm2d1 Adgrf1 Calcrl
44 54 35 111 162 107 36 58 124 126 133 146 153 122 188
8 Gpr160 Adgre5 Adra2b Tm2d1 F2rl1 P2ry2 Gprc5c P2ry2 Gpr160 Gpr160 Gcgr Ackr3 Adora1 Adora1 Adgrl4
41 52 32 102 106 102 36 54 108 104 129 136 150 96 159
9 Sucnr1 Adra2b Smo Fzd4 Fzd4 Gpr137 Adgra3 Gpr160 Fzd4 Fzd4 Ptgfr Tm2d1 Ackr3 Gprc5b Tbxa2r
38 49 30 86 99 71 35 49 90 85 125 125 118 65 152
10 Adra2b Adrb2 Adgra3 F2r P2ry2 Adgrl4 Fzd4 Gpr137 Adgra3 Avpr1a Gpr160 Gpr160 Fzd4 Gpr137 Ccrl2
34 39 27 85 89 59 31 46 81 52 120 118 84 60 134

For all G protein-coupled receptor data, entries were sorted based on maximum expression (in transcripts per kilobase million) in each individual tubule segment. ATL, thin ascending limb of the loop of Henle; CCD, cortical collecting duct; CNT, connecting tubule; CTAL, cortical thick ascending limb of the loop of Henle; DCT, distal convoluted tubule; DTL1, short descending limb of the loop of Henle; DTL2, long descending limb of the loop of Henle in the outer medulla; DTL3, long descending limb of the loop of Henle in the inner medulla; Glom, glomerulus; IMCD, inner medullary collecting duct; MTAL, medullary thick ascending limb of the loop of Henle; PTS1, initial segment of the proximal convoluted tubule; PTS2, proximal straight tubule in cortical medullary rays; PTS3, last segment of the proximal straight tubule in the outer stripe of outer medulla; OMCD, outer medullary collecting duct.

These GPCRs were matched to known ligands, coupled G proteins, and known physiological roles in the segment in which it is selectively expressed. For ease of reference, principal pathways known to be activated by each G protein α-subunits are shown in Table 2. Although we primarily focused on the downstream effects of G α-subunits, it is worth noting that other heterotrimeric G protein subunits, Gβγ complexes, have been established to have downstream signaling effects, which have been the subject of detailed reviews (19, 20). In addition, β-arrestins have been implicated to have additional effects downstream of GPCR signaling (21). For example, established renal GPCRs such as PTH 1 receptor (Pth1r) and arginine vasopressin V2 receptor (Avpr2) have been found to have noncanonical signaling pathways via β-arrestins (22, 23). The focus of this review is on these selectively expressed GPCRs to identify understudied GPCRs with specific patterns of expression from which, given our current understanding of transport physiology, hypotheses and future studies could be proposed.

Table 2.

G protein α-subunits

G Protein α-Subunit Downstream Effects
Gαs Activate adenylyl cyclase → increase cAMP → activate PKA
Gαi/Gαo Inhibit adenylyl cyclase → decrease cAMP
Gαq/Gα11 Activate phospholipase Cβ: IP3 generation → intracellular Ca2+ and diacylglycerol → protein kinase C
Gα12/Gα13 RhoGEF → RhoA → activate Rho kinase

Shown is a summary of G protein α-subunits that bind to G protein-coupled receptors and their general downstream effects. IP3, inositol 1,4,5-trisphosphate; RhoGEF; rho-specific guanine nucleotide exchange.

GPCRs SELECTIVELY EXPRESSED ACROSS THE NEPHRON

Proximal Tubule

GPCRs that are selectively expressed in the PT, microdissected as three segments (PTS1, PTS2, and PTS3), are shown in Fig. 2. Known ligands, G proteins, and physiological roles for PT-selective GPCRs are shown in Table 3. This list includes multiple well-studied receptors that are known to localize to this segment, including Pth1r, angiotensin II (ANG II) receptor type 1a (Agtr1a), two α-adrenergic receptors (Adra1a and Adra2b), and succinate receptor 1 (Sucnr1).

Figure 2.

Figure 2.

Proximal tubule-selective G protein-coupled receptors (GPCRs). GPCR expression data from microdissected renal tubules were filtered based on an individual genes’ maximum expression [in transcripts per kilobase million (TPM)] in the initial segment of the proximal convoluted tubule (PTS1), proximal straight tubule in cortical medullary rays (PTS2), or last segment of the proximal straight tubule in the outer stripe of outer medulla (PTS3) compared with its expression in any other tubule segment. PTS1, PTS2, and PTS3 expression that was 1.5-fold greater than any other tubule segment was considered selective. Entries are sorted starting with the greatest TPM ratio. Refer to Table 3 for additional information about each GPCR. ATL, thin ascending limb of the loop of Henle; CCD, cortical collecting duct; CNT, connecting tubule; CTAL, cortical thick ascending limb of the loop of Henle; DCT, distal convoluted tubule; DTL1, short descending limb of the loop of Henle; DTL2, long descending limb of the loop of Henle in the outer medulla; DTL3, long descending limb of the loop of Henle in the inner medulla; Glom, glomerulus; IMCD, inner medullary collecting duct; MTAL, medullary thick ascending limb of the loop of Henle; OMCD, outer medullary collecting duct.

Table 3.

Proximal tubule-selective G protein-coupled receptors

Gene Symbol Annotation Ligand G Protein Renal Physiological Process Key Reference(s)
Lpar3 Lysophosphatidic acid receptor 3 Lysophosphatidic acid Gαi/Gαq Segment function not known. Blockade reduces ischemia-reperfusion injury (24)
Cckar Cholecystokinin A receptor Cholecystokinin Gαq/Gαs Segment function not known. Proposed role in the natriuretic response (25)
Sucnr1 Succinate receptor 1 Succinic Acid Gαi/Gαq Succinate binding induces renin release (26, 27)
Agtr1a angiotensin II receptor, type 1a Angiotensin II Gαi/Gαo/ Gαq Na+/K+-ATPase activity, HCO3 HCO3 reabsorption, renin-angiotensin system activation (2831)
Adra2b Adrenergic receptor, α2b Epinephrine/norepinephrine Gαi/Gαo MAPK activation, Na+-K+-ATPase activity (3234)
Adgrg4 Adhesion G protein-coupled receptor G4 Orphan   Kidney function not known
Adra1a Adrenergic receptor, α1a Epinephrine/norepinephrine Gαq/Gα11 Na+ absorption (35, 36)
Lpar6 Lysophosphatidic acid receptor 6 Lysophosphatidic acid Gαs/Gαi/ Gα12 Segment function not known
Pth1r Parathyroid hormone 1 receptor Parathyroid hormone Gαs Inhibits phosphate/bicarbonate absorption (5, 3739)

Shown are known ligands, coupled G protein α-subunits, and segment-specific physiological roles for G protein-coupled receptors selectively expressed in the proximal tubule.

Pth1r localizes to the PT (37, 40, 41). Classic isolated perfused tubule studies have demonstrated that PTH acting on the basolateral membrane strongly inhibits salt and water reabsorption (38, 42, 43) through effects to increase adenylyl cyclase activity (3) by activation of Gαs (39).

Agtr1a also has established functions in the PT (30, 44, 45). Activation of this receptor by ANG II occupation results in acceleration of salt, glucose, and water reabsorption (29, 46, 47) through effects to decrease cAMP production (46, 47), consistent with its known coupling to Gαi (28). Notably, expression of Agtr1a is much lower in segments beyond the PT, a pattern confirmed in other single-cell RNA-seq studies (14).

The data shown in Fig. 2 also indicate that the PT selectively expresses two α-adrenergic receptors, α-1A (Adra1a) and α-2B (Adra2b), in agreement with the literature (32, 34, 36). Activation of α-adrenergic signaling increases Na+ and bicarbonate reabsorption in the PT by inhibiting cAMP (4850). α-1A couples to Gαq/Gα11 (35), whereas α-2B couples to Gαi/Gαo (33). We note that the expression of multiple other adrenergic receptors across the nephron and will go into further detail on this GPCR subfamily later on.

Finally, Fig. 2 shows that Sucnr1 (also known as Gpr91) is selectively expressed in the PT, corroborating prior findings (26). Succinate receptor 1 activation leads to increasing Ca2+ mobilization and inhibition of cAMP via Gαi or Gαq coupling (26). A recent study has suggested that Gαi-induced Ca2+ signals are dependent on a separate Gαq-coupled signaling cascade (51). It’s possible that succinate receptor 1 signals in a similar fashion. Although normal physiological levels of extracellular succinate are insufficient to activate succinate receptor 1 (26, 27), plasma succinate is known to increase in animal disease models of diabetes (27), hypertension (52), and liver damage (53). The physiological role of succinate receptor 1 activation in the PT remains incompletely characterized.

In addition to GPCRs whose expression is consistent with prior studies, the unbiased nature of omics approaches reveal expression of receptors that have not been well explored in renal physiology. For example, the cholecystokinin (CCK) receptor (Cckar) is selectively expressed in the PT. CCK is a digestive regulatory hormone secreted by the proximal intestine, and Cckar is also highly expressed in the stomach, pancreas, and gastrointestinal smooth muscle (54). Cckar PT expression has been previously reported in mice and humans (25, 55, 56), but its function has not been fully explored. Of note, Takahashi-Iwanaga et al. (25) also reported the expression of CCK in renal medullary cells, which secrete CCK in response to body fluid volume expansion. This work suggests that CCKAR could be part of a feedback loop in response to Na+ and fluid balance, although these studies did not examine the effect of CCK on PT NaCl and water absorption. RNA-seq confirms that CCKAR ligand CCK is expressed in several medullary segments, namely, the descending thin limb of Henle’s loop (DTL), ascending thin limb of Henle’s loop (ATL), medullary TAL (MTAL), and inner medullary CD (IMCD) (9). Future studies involving treatment of isolated perfused PTs with CCK and measurement of transport would be a key experiment in uncovering the role of Cckar in kidney function.

Also present in the PT are adhesion G protein-coupled receptor G4 (Adgrg4), an orphan receptor, as well as lysophosphatidic acid (LPA) receptor 3/6 (Lpar3/6). Other receptors in these subfamilies are expressed in other nephron segments, and we go into more detail about trends in their expression in expression of gpcr families in the renal tubule.

There has also been increasing evidence in recent years of sex differences in kidney function, including transport regulation (57). In a single-cell RNA-seq study by Ransick et al. (14), clustering of renal tubule cells by gene expression revealed separation by sex, although only in the PT. Multiple GPCRs identified in our data as being selective for the PT also exhibited sex differences in expression, as shown in Fig. 3. Sucnr1 and Cckar, as previously described, were more highly expressed in males compared with females. Adra1a, one of multiple adrenergic receptors selectively expressed in the renal tubule, was more highly expressed in females. Future studies will be needed to determine if these GPCR sex differences directly translate to differences in tubule function and if these differences are relevant in human tissue.

Figure 3.

Figure 3.

Sex differences in G protein-coupled receptor (GPCR) expression in the proximal tubule. Expression data from Ransick et al. (14) were compared between males and females in the initial segment of the proximal convoluted tubule (PTS1; top) and proximal straight tubule in cortical medullary rays (PTS2; bottom). Fold changes in gene expression [male/female (M/F)] for all detected GPCRs were averaged, and a 95% confidence interval was calculated. Shown here are all GPCRs outside this confidence interval. GPCRs preferentially expressed in males are shown in blue; GPCRs preferentially expressed in females are shown in red. [Adapted from data published in Ransick et al. (14).]

Thin limbs of Henle’s loop.

Functional studies in the isolated DTL and ATL are technically challenging, and so very few GPCRs have had their functions linked directly to this nephron region. GPCRs that are selectively expressed in the DTL (DTL1, DTL2, and DTL3) and ATL are provided in Supplemental Fig. S1 (all Supplemental Material is available at https://hpcwebapps.cit.nih.gov/ESBL/Database/GPCRSupplemental/). What is known about the ligands and physiological roles for receptors in the DTL and ATL is outlined in Supplemental Table S1.

TAL of Henle’s loop.

GPCRs that are selectively expressed in the TAL are shown in Fig. 4. Known ligands and physiological roles for these receptors are shown in Table 4. Four receptors were identified as being selectively expressed in the MTAL and/or cortical thick ascending limb, namely, the prostaglandin E2 EP3 receptor (Ptger3) (58), Ca2+-sensing receptor (Casr) (61, 63), glucagon receptor (Gcgr) (6), and opsin 3 (Opn3). These receptors are also expressed at lower levels in the distal convoluted tubule (DCT), connecting tubule (CNT), and cortical CD (CCD). Ptger3, Casr, and Gcgr are all well-established TAL GPCRs. As elucidated by the classic studies by Morel and coworkers (3, 4, 64), Glucagon receptor along with the vasopressin and PTH receptor activate adenylyl cyclase in TAL cells consistent with signaling via Gαs (Table 4). Recent studies have corroborated these findings (6062). Isolated perfused tubule studies have demonstrated that basolateral addition of vasopressin, PTH, and calcitonin increase NaCl absorption in the TAL (6569). In contrast, prostaglandin E2 inhibits NaCl absorption in the TAL, counteracting the stimulatory effect of vasopressin (67, 70). Activation of the EP3 receptor inhibits basolateral K+ channels, causing depolarization and thereby inhibiting NaCl reabsorption (59). Signaling by the EP3 receptor occurs via Gαi (Table 4). The Ca2+-sensing receptor is Gαi/Gαq coupled and inhibits voltage-dependent Ca2+ transport/reabsorption in the TAL (61).

Figure 4.

Figure 4.

Thick ascending limb-selective G protein-coupled receptor (GPCRs). GPCR expression data from microdissected renal tubules were filtered based on an individual genes’ maximum expression [in transcripts per kilobase million (TPM)] in either the cortical thick ascending limb (CTAL) or medullary thick ascending limb (MTAL) compared with its expression in any other tubule segment. MTAL/CTAL expression that was 1.5-fold greater than any other tubule segment was considered selective. Entries are sorted starting with the greatest TPM ratio. Refer to Table 4 for additional information about each GPCR. ATL, thin ascending limb of the loop of Henle; CCD, cortical collecting duct; CNT, connecting tubule; DCT, distal convoluted tubule; DTL1, short descending limb of the loop of Henle; DTL2, long descending limb of the loop of Henle in the outer medulla; DTL3, long descending limb of the loop of Henle in the inner medulla; Glom, glomerulus; IMCD, inner medullary collecting duct; PTS1, initial segment of the proximal convoluted tubule; PTS2, proximal straight tubule in cortical medullary rays; PTS3, last segment of the proximal straight tubule in the outer stripe of outer medulla; OMCD, outer medullary collecting duct.

Table 4.

Thick ascending limb-selective G protein-coupled receptors

Gene Symbol Annotation Ligand G Protein Renal Physiological Process Key References
Ptger3 Prostaglandin E receptor 3 (subtype EP3) Prostaglandin E2 Gαi Inhibits NaCl reabsorption (58, 59)
Casr Ca2+-sensing receptor Ca2+ Gαi/Gαq Inhibits Ca2+ reabsorption (60, 61)
Gcgr Glucagon receptor Glucagon Gαs Stimulates adenylyl cyclase activity (4, 6, 62)
Opn3 Opsin 3 Chromophore Unknown Kidney function not known

Shown are known ligands, coupled G protein α-subunits, and segment-specific physiological roles for G protein-coupled receptors selectively expressed in the thick ascending limb.

Opn3 is also expressed in the TAL and at lower levels in the DCT, CNT, and CCD. Opn3 has been characterized in brain, skin, and adipose tissue, but no functional studies have been conducted on Opn3 in the kidney (7173). Studies have shown that opsin 3 can be light sensitive like rhodopsin, yet is expressed in tissues that are not exposed to light and has downstream functions that are not activated by light, suggesting light-independent mechanisms of opsin activation (71). These studies have yielded tools, such as a universal Opn3 reporter mouse, that could be valuable in kidney studies of this receptor.

Macula densa.

Macula densa cells are neuroepithelial-like cells that sense luminal NaCl concentration, providing a feedback signal that regulates renin secretion and tubuloglomerular feedback (74). The transcriptome of macula densa cells has recently been elucidated by single-cell RNA-seq studies in the mouse (75). These studies confirmed the expression of the arginine vasopressin 1a (V1a) receptor (76), where it mediates an indirect action to increase renin secretion. In addition, it identified two other GPCRs in macula densa cells, namely, adhesion G protein-coupled receptor F1 (Adgrf1) and thyroid stimulating hormone receptor (Tshr). The latter has previously been shown to be expressed in the kidney (77), although not specifically in the macula densa. Aside from thyroid stimulating hormone, it also acts as a receptor for the heterodimeric glycoprotein hormone GPHA2-GPHB5.

DCT and CNT.

GPCRs selectively expressed in the DCT and CNT are shown in Fig. 5. Known ligands and physiological roles for these receptors are shown in Table 5. Both the adenosine a2b receptor (Adora2b) and prostaglandin F receptor (Ptgfr) are selectively expressed in the DCT, although the latter has significant levels of expression extending into the CD. This pattern of expression is in agreement with other surveys of these receptors (79, 81).

Figure 5.

Figure 5.

Distal convoluted tubule (DCT)- and connecting tubule (CNT)-selective G protein-coupled receptor (GPCRs). GPCR expression data from microdissected renal tubules were filtered based on an individual genes’ maximum expression [in transcripts per kilobase million (TPM)] in either the DCT or CNT compared with its expression in any other tubule segment. DCT/CNT expression that was 1.5-fold greater than any other tubule segment was considered selective. Entries are sorted starting with the greatest TPM ratio. Refer to Table 5 for additional information about each GPCR. ATL, thin ascending limb of the loop of Henle; CCD, cortical collecting duct; CTAL, cortical thick ascending limb of the loop of Henle; DTL1, short descending limb of the loop of Henle; DTL2, long descending limb of the loop of Henle in the outer medulla; DTL3, long descending limb of the loop of Henle in the inner medulla; Glom, glomerulus; IMCD, inner medullary collecting duct; MTAL, medullary thick ascending limb of the loop of Henle; PTS1, initial segment of the proximal convoluted tubule; PTS2, proximal straight tubule in cortical medullary rays; PTS3, last segment of the proximal straight tubule in the outer stripe of outer medulla; OMCD, outer medullary collecting duct.

Table 5.

Distal convoluted tubule- and connecting tubule-selective G protein-coupled receptors

Gene Symbol Annotation Ligand G Protein Cell Type Renal Physiological Process Key References
Distal convoluted tubule
Adora2b Adenosine A2b receptor Adenosine Gαs   Induce H secretion via V-ATPase (7880)
Ptgfr Prostaglandin F receptor Prostaglandin F Gαq/Gα11 Principal cells Modulates K channels (8183)
Connecting tubule
Sctr Secretin receptor Secretin Gαs β-Intercalated cells > α-intercalated cells Segment function not known (84, 85)
Fzd7 Frizzled class receptor 7 Wnt Gαs/Gαi Also signals through Wnt, segment function not known
Oxgr1 oxoglutarate receptor 1 α-ketoglutarate Gαq/Gα11 α-Intercalated cells > β-intercalated cells > principal cells Segment function not known (26, 86)

Known ligands, coupled G protein α-subunits, cell type-specific expression, and segment-specific physiological roles for G protein-coupled receptors selectively expressed in the distal convoluted tubule and connecting tubule are shown.

The effect of ADORA2B activation on DCT transport has not been studied as of this writing. However, activation of ADORA2B in medullary α-intercalated cells induced cAMP/PKA signaling to increase V-ATPase translocation, consistent with signaling through Gαs (78). In the context of hypertension, elevated levels of renal adenosine led to increased Adora2b expression, increasing hypoxia-inducible factor-1α and endothelin-1 (80), although these effects have not been linked to a particular tubule segment.

Prostaglandin F receptor has been shown to modulate K+ channels in the DCT (82). Its ligand, prostaglandin F, is formed from prostaglandin H2, catalyzed by the ubiquitous enzyme aldose reductase (AKR1B1/AKR1B3), which is also involved in osmotic regulation in the kidney (87). A recent study has shown that activation of PTGFR in the DCT stimulates Cl channel ClC-K2 (Clcknb) and NaCl cotransporter (NCC; Slc12a3) via protein kinase C and p38 activation (83). Ptgfr likely signals via Gαq (88, 89). Both CIC-K2 and NCC are also stimulated in models of salt-sensitive hypertension; the role of Ptgfr in this context may be of interest for future translational studies (31).

As shown in Fig. 5, the secretin receptor (Sctr) is also expressed in the CNT and to a lesser extent in the CCD. Single-cell RNA-seq studies (75) have shown expression of Sctr mRNA in both α- and β-intercalated cells but not in CNT cells, suggesting a role in acid-base transport. Similar single-cell RNA-seq studies of CD cell types also showed Sctr expression in intercalated cells but not in principal cells (12). Consistent with these results, early studies have shown that secretin can induce urinary alkalinization via increased HCO3 excretion (90). In isolated perfused tubules, secretin treatment stimulated HCO3 secretion via β-intercalated cells, which was significantly reduced in tubules from mice lacking cystic fibrosis transmembrane regulator (84). Secretin has also been proposed to increase water reabsorption in aquaporin-2-expressing segments, suggesting a role in CNT cells, principal cells, and/or IMCD cells (85), contrasting with the apparent absence of Sctr mRNA in aquaporin-2-expressing cells (12, 14, 75). Treatment with secretin alone or in conjunction with other compounds has been shown to ameliorate polyuria seen in X-linked nephrogenic diabetes insipidus mouse models (91). However, whether or not secretin can act as a true substitute for vasopressin by increasing water permeability remains an open question. Direct measurements of water permeability responses to secretin in isolated, perfused CDs or with in vivo perfusion of CNT segments will be required to determine whether observed effects of secretin on water excretion are direct or indirect.

As shown in Fig. 5, oxoglutarate receptor 1 (Oxgr1) is also highly expressed in the CNT and to a lesser extent in the DCT and CD, as seen in a prior study (26). Single-cell RNA-seq studies have indicated that Oxgr1 mRNA is expressed in intercalated cells, principal cells, and CNT cells, but at several-fold higher levels in intercalated cells (both α and β subtypes). In isolated perfused CCD experiments, treatment with oxoglutarate led to activation of OXGR1 to stimulate Cl reabsorption and bicarbonate secretion in a process mediated by the Cl/bicarbonate exchanger pendrin (86, 92). Notably, these effects are mediated by apically localized OXGR1 in intercalated cells (92). The dependence of this effect on protein kinase C is consistent with OXGR1 signaling through Gαq (86). These effects are attributable to Oxgr1 in β-intercalated cells. Because Oxgr1 is also expressed in the other cell types of the CNT/CCD (12, 14), it seems possible that oxoglutarate has additional pendrin-independent functions in the CNT/CCD.

The Wnt receptor Frizzled class receptor 7 (Fzd7) is selectively expressed in the CNT and to a lesser extent in the DCT and CCD. Although Frizzled receptors do not typically couple to G proteins, FZD7 has been found to couple to Gαs or Gαi in different contexts (93). Canonical Wnt signaling is highly upregulated during kidney development, in particular as the ureteric bud differentiates into the CD system (9496). Although this pathway is generally downregulated in adult tissue, upregulated Wnt signaling has been observed in chronic kidney disease and kidney injury (9799). The role of Fzd7 in the adult kidney CNT is currently unknown (100).

Collecting duct.

GPCRs selectively expressed in the CD are shown in Fig. 6. This includes the CCD, outer medullary CD (OMCD), and IMCD. Known ligands and physiological roles for these receptors are shown in Table 6. GPCRs in this segment include the well-characterized receptors Avpr2 and prostaglandin E receptor 1 (Ptger1). In isolated perfused tubule studies, Grantham and Orloff (108) demonstrated that vasopressin increases osmotic water permeability in rabbit CCDs and that this action was mimicked by the addition of cAMP analogs and cyclic nucleotide phosphodiesterase inhibition. Later, Imbert et al. (7) demonstrated that vasopressin activates adenylyl cyclase in microdissected CDs. Although two vasopressin receptors are expressed in the CD, Avpr2 is the only one expressed in principal cells and IMCD cells, whereas the arginine vasopressin 1a receptor is abundant in α- and β-intercalated cells (12). Among the three vasopressin receptors (V1a, V1b, and V2), the V2 receptor is the only one coupled to Gαs (107). In recent years, extensive studies, including those in our laboratory, have demonstrated features of V2 receptor action in the CD. Aside from the well-studied effect of vasopressin action through the V2 receptor to speed water transport across the CD epithelium, vasopressin also increases urea transport via urea transporter (UT)-A1 and UT-A3 urea channels (111113) and increases Na+ transport (114, 115) through regulation of β- and γ-epithelial Na+ channel subunits (116119).

Figure 6.

Figure 6.

Collecting duct (CD)-selective G protein-coupled receptor (GPCRs). GPCR expression data from microdissected renal tubules were filtered based on an individual genes’ maximum expression [in transcripts per kilobase million (TPM)] in either the cortical CD (CCD), outer medullary CD (OMCD), or inner medullary CD (IMCD) compared with its expression in any other tubule segment. CCD/OMCD/IMCD expression that was 1.5-fold greater than any other tubule segment was considered selective. Entries are sorted starting with the greatest TPM ratio. Refer to Table 6 for additional information about each GPCR. ATL, thin ascending limb of the loop of Henle; CNT, connecting tubule; CTAL, cortical thick ascending limb of the loop of Henle; DCT, distal convoluted tubule; DTL1, short descending limb of the loop of Henle; DTL2, long descending limb of the loop of Henle in the outer medulla; DTL3, long descending limb of the loop of Henle in the inner medulla; Glom, glomerulus; MTAL, medullary thick ascending limb of the loop of Henle; PTS1, initial segment of the proximal convoluted tubule; PTS2, proximal straight tubule in cortical medullary rays; PTS3, last segment of the proximal straight tubule in the outer stripe of outer medulla.

Table 6.

Collecting duct-selective G protein-coupled receptors

Gene Symbol Annotation Ligand G Protein Cell Type Renal Physiological Process Key Reference(s)
Adgrf1 Adhesion G protein-coupled receptor F1 Orphan Kidney function not known
Ptger1 Prostaglandin E receptor 1 Prostaglandin E2 Gαq/Gα11 Principal cells Inhibits Na+ absorption (101, 102)
P2ry14 Purinergic receptor P2Y, G protein coupled, 14 UDP-glucose Gαi α-Intercalated cells Inflammatory response (103, 104)
Gprc5b G protein-coupled receptor, family C, group 5, member B Orphan Principal cells Kidney function not known
Adgrf5 Adhesion G protein-coupled receptor F5 Tethered agonist Gαq/Gα11 α-Intercalated cells V-ATPase trafficking (105)
Npy6r Neuropeptide Y receptor Y6 Neuropeptide Y/peptide YY/pancreatic peptide Principal cells Segment function not known, pseudogene in humans (106)
Fzd1 Frizzled class receptor 1 Wnt Gαi/Gαq Principal cells Also signals through Wnt
Npy1r Neuropeptide Y receptor Y1 Neuropeptide Y/peptide YY Gαi/Gαo Kidney function not known (106)
Avpr2 Arginine vasopressin receptor 2 Arginine vasopressin Gαs Principal cells Water reabsorption (7, 107, 108)
Celsr2 Cadherin, EGF LAG seven-pass G-type receptor 2 Orphan Principal cells > β-intercalated cells > α-intercalated cells
Lpar1 Lysophosphatidic acid receptor 1 Lysophosphatidic acid Gαi/Gαq/Gα12 Principal cells Segment function not known
Gper1 G protein-coupled estrogen receptor 1 17β-Estradiol Gαi/Gαo Principal cells Ca2+-H+-ATPase (109)
S1pr3 Sphingosine-1-phosphate receptor 3 Sphingosine-1 phosphate Gαi/Gαq β-Intercalated cells Kidney function not known (110)

Known ligands, coupled G protein α-subunits, cell type-specific expression, and segment-specific physiological roles for G protein-coupled receptors selectively expressed in the collecting duct are shown.

As shown in Fig. 6, Ptger1, whose receptor is linked to Gαq or Gα11, is also expressed in CD cells (101, 102). Activation of this receptor inhibits water and NaCl reabsorption (102). In the CD, Ptger1 is expressed predominantly in principal cells rather than intercalated cells (12).

Frizzled class receptor 1 (Fzd1) is also selectively expressed in CD principal cells (12). A key component of the canonical Wnt signaling pathway, FZD1 binds Wnt along with coreceptors low-density lipoprotein receptor-related proteins 5 and 6 (LPR5 and LPR6, respectively). Single-cell RNA-seq studies have shown that Lpr6 is also selectively expressed in principal cells (120). As previously described, Wnt signaling plays key roles in kidney development; however, the role of Fzd1 in the adult kidney CD has not been established. In addition to its role in canonical Wnt signaling, FZD1 has also been reported to couple to either Gαi or Gαq in different contexts (93, 121, 122).

Also of interest is the purinergic receptor P2Y14 (P2rY14 gene; Fig. 6), which has been identified as being selectively expressed in α-intercalated cells (12, 103). Work by Azroyan et al. (103) has shown that activation of this receptor via UDP-glucose induces expression of proinflammatory cytokines, suggesting that purinergic receptors in α-intercalated cells are key to the regulation of inflammation in the CD. This was followed up in a study by Battistone et al. (104), who demonstrated that ablation of P2rY14 prevented the proinflammatory chemokine response after ischemia-reperfusion injury and reduced kidney damage, reinforcing this receptor’s role in the proinflammatory response. In vitro kidney cell models suggest that P2Y14 couples to Gαi, and activation leads to downstream ERK1/2 phosphorylation in human embryonic kidney and C6 cell lines, although this has not been verified in vivo (123).

As shown in Fig. 6, two neuropeptide Y (NPY) receptors, Npy1r and Npy6r, are selectively expressed in CD segments of the mouse, consistent with prior work (106). Npy6r is truncated in humans (124), where it is considered a pseudogene. NPY1R can bind a variety of ligands, including NPY, peptide YY (PYY), and pancreatic polypeptide (PP) (125, 126). Classically, NPY is secreted by renal adrenergic nerve fibers cosecreted with norepinephrine. PYY is secreted by the small intestine in response to feeding (127), and PP is secreted by γ-cells of pancreatic islets (128). Interestingly, expression of the NPY1R/NPY6R ligand NPY has been localized to the renal tubule (129), and our RNA-seq data indicate that NPY is expressed in both the DTL and OMCD, suggesting local activation of NPY receptors (9). NPY1R is coupled to Gαi, suggesting that it should inhibit cAMP production and decrease water permeability and active Na+ transport. Indeed, NPY was found to inhibit vasopressin-stimulated osmotic water permeability in isolated CDs (130). NPY signaling has been implicated in renal vasoconstriction and renin release, although these effects have not been linked to any NPY receptor activity in the CD (131).

Less well-established receptors selectively expressed in the CD (Fig. 6) include G protein-coupled estrogen receptor 1 (Gper1), sphingosine-1-phosphate receptor 3 (S1pr3), Adgrf1, adhesion G protein-coupled receptor F5 (Adgrf5), cadherin EGF Lag seven-pass G-type receptor 2 (Celsr2), and lysophosphatidic acid receptor 1 (Lpar1).

Most actions of estrogens are mediated by nuclear receptors that act as transcription factors, directly regulating transcription of target genes. However, a GPCR known as GPER1 or GPR30 has been identified that binds steroids, including estradiol (132, 133), and triggers intracellular Ca2+ mobilization and synthesis of phosphatidylinositol 3,4,5-trisphosphate (134). Prior studies have identified Gper1 in microdissected DCT, CNT, and CCD segments (109, 135), matching the distribution found by RNA-seq (Fig. 6). These tubule segments manifested 17β-estradiol-induced increases in Ca2+ concentration (109) consistent with GPER1 activation. Although the authors concluded that the site of Gper1 actions was in intercalated cells, recent single-cell RNA-seq analysis has identified Gper1 expression in CD principal cells, CNT cells, and DCT cells without a significant signal in intercalated cells (14). Consistent with a DCT cell site of action, GPER1 has been found to regulate thiazide-sensitive NCC of the DCT by binding aldosterone (136). Activation of GPER1 in the renal medulla has been found to cause natriuresis (selectively in female rats) via endothelin type A and B receptors, pointing to a physiological role of GPER1 in the regulation of Na+ and Cl excretion (137). The endothelin type B receptor is strongly expressed in the DCT and all CD segments (12).

S1pr3 is also expressed in the CD (Fig. 6). Although a function for this receptor has not been described in the CD, the functions of this receptor are well established in antigen-presenting cells in that kidney, which respond to sphingosine-1 phosphate to regulate inflammatory responses and immune cell migration (138). S1pr3 knockout mice have been described as being protected from kidney ischemia-reperfusion injury (110). S1pr3 is thought to be Gαi/Gαq coupled (139).

Of note, the most specific CD GPCR is Adgrf1, which has been shown to bind synaptamide (a DHA metabolite) in the fetal brain (140), although its function has not been explored in the kidney. In addition, Adgrf5, Celsr2, and Lpar1 are also expressed in the CD. These receptors belong to the adhesion class and lysophosphatidic acid families of GPCRs, which have multiple members expressed along the tubule and will be discussed in more detail later on. Finally, mRNA and protein expression of orphan receptor GPCR class C group 5 member B has been observed in the CD. Despite being highly expressed in the kidney CD, very little is known about this orphan receptor. Further studies are needed to uncover the function of these specific and highly expressed GPCRs.

EXPRESSION OF GPCR FAMILIES IN THE RENAL TUBULE

Adhesion GPCRs

Although we observe specific expression of well-characterized individual receptors, there are trends in multiple GPCR families expressed across the nephron. Expression of GPCR groups as well as specific localization can give us hints as to functional roles for these receptors. RNA-seq analysis has identified multiple members of the family adhesion GPCRs (aGPCRs) that are selectively expressed in tubule segments, including Adgrg4 in the PT, adhesion G protein-coupled receptors L3 and L4 (Adgrl3 and Adgrl4, respectively) in the DTL, adhesion G protein-coupled receptor E5 (Adgre5) in the ATL, and Adgrf1, Adgrf5, and Celsr2 in the CD (Fig. 7). Nine other members of this gene family are expressed more universally, including Adgrg1, by far the most highly expressed aGPCR in the nephron. Adgrg1 expression, along with Adgrf5 and adhesion G protein-coupled receptor L2 (Adgrl2), has been confirmed in rat proteomic studies (16). The role of aGPCRs in physiology and disease has been the subject of multiple reviews that provide a more detailed picture of our current understanding of this gene family (141143).

Figure 7.

Figure 7.

Expression of adhesion G protein-coupled receptors along the renal tubule. Colors indicate segment boundaries. CD, collecting duct; DCT, distal convoluted tubule; DTL, descending thin limb; Glom, glomerulus; PT, proximal tubule; TAL, thick ascending limb.

aGPCRs are characterized by their long extracellular domains, which contain a GPCR autoproteolysis-inducing (GAIN) domain, cleavage of which exposes a tethered ligand capable of activating the receptor (143, 144). However, aGPCRs are capable of being activated by multiple mechanisms, including exogenous ligands, mechanical stimuli, and cell-cell interactions with other external membrane proteins (145148). ADGRG1, for example, can be activated by collagen type III (146), which is upregulated in renal fibrosis (149), and l-phenylalanine, although only at high levels (145).

To date, few aGPCR functions have been directly linked to the functions of specific kidney cell types, and many receptors remain orphans. Experimental progress has been challenged by a lack of validated antibodies and molecular tools (141). In addition, multiple knockout models for aGPCRs have been generated that have no overt renal phenotypes, suggesting redundant aGPCR functions (150, 151), although deeper analysis of renal function may yet identify functional roles. For example, mice lacking adhesion G protein-coupled receptor G3 (Adgrg3), which is expressed in the DTL and CD (9), were partially protected during acute kidney injury (152). The crystal structure of ADGRG3 as well as its binding to glucocorticoid ligands have also recently been published and may yield insights into the structure and binding properties of other aGPCRs (153). In addition, a recent study by Zaidman et al. (105) found a specific role for Adgrf5 (Gpr116) in α-intercalated cells of the CD, the cells normally responsible for urinary acidification through proton secretion (154). Adgrf5 knockout mice had acidified urine and metabolic alkalosis indicative of a static role of ADGRF5 to inhibit proton secretion, which appears to be mediated by regulation of trafficking of vacuolar H+-ATPase to the cell surface (105). The functions of many other aGPCRs expressed along the renal tubule are yet to be uncovered.

Adrenergic Receptors

Renal sympathetic nerve activity plays key roles in blood pressure regulation in part through control of Na+ and water reabsorption along the renal tubule (155). These effects are mediated by binding of circulating epinephrine and norepinephrine to adrenergic receptors. Multiple members of the adrenergic receptor family are highly and selectively expressed in distinct renal tubule segments (Fig. 8). Adrenergic receptors can be divided into three groups, roughly in accord with the heterotrimeric G protein α-subunit that they signal through, viz. α1-adrenergic receptors through Gαq/11, α2-adrenergic receptors through Gαi, and β-adrenergic receptors through Gαs. PTs express all three, although in different distributions (Fig. 8). In isolated proximal convoluted tubules, norepinephrine was found to strongly increase NaCl and fluid absorption (156), seemingly consistent with a role in Na+ balance and blood pressure regulation. The finding that α2-adrenergic receptors counter the increase in cAMP in PTs is consistent with a potential role of these receptors in stimulation of NaCl transport (50), which occurs through an increase in apical Na+/H+ exchange (157). RNA-seq results (Fig. 8) have identified Adra2b as the receptor most likely to mediate this effect. In contrast, proximal α1-adrenergic receptors stimulate gluconeogenesis (158), and this action is likely to be restricted to the S-1 PT based on the RNA-seq finding that Adra1a is only expressed in S-1. In isolated perfused proximal S-2 segments, norepinephrine had no effect on NaCl and fluid absorption (156), perhaps because this segment expresses both β2- and α2b-adrenergic receptors (Fig. 8), which have opposing effects on cAMP production.

Figure 8.

Figure 8.

Expression of adrenergic receptors along the renal tubule. Colors indicate segment boundaries. CD, collecting duct; DCT, distal convoluted tubule; Glom, glomerulus; DTL, descending thin limb; PT, proximal tubule; TAL, thick ascending limb.

Besides the PT, additional renal tubular sites of catecholamine-mediated regulation of salt and water transport are the CD and DCT. In the CCD, α2-adrenergic receptor activation has been shown to oppose vasopressin action by decreasing cAMP (159, 160). Isolated perfused tubule experiments in CCDs showed that the α-adrenergic agonist phenylephrine markedly inhibited the ability of vasopressin to stimulate fluid absorption and osmotic water permeability (161). The RNA-seq results showed that the α-adrenergic receptor expressed in the CD is α2a (Adra2a; Fig. 8), i.e., a different α2-receptor than that expressed in the PT. Inhibition of Na+ and water absorption by adrenergic agents in the CD would be expected to decrease blood pressure and would tend to oppose α2b-adrenergic receptor effects in the PT to increase Na+ and water absorption. As shown in Fig. 8, β-adrenergic receptors (Adrb2) are also expressed in CD segments. However, a single-cell RNA-seq study (12) has indicated that CD Adrb2 expression is exclusively in intercalated cells. Consistent with this, β-adrenergic agonists increased Cl absorption in isolated perfused CCDs without affecting net Na+ transport or water permeability (162).

β2-Adrenergic receptors are also expressed in the DCT. Norepinephrine, acting through β-adrenergic receptors, has been demonstrated to increase phosphorylation of NCC of the DCT at a site demonstrated to be associated with increased transport activity (163). This effect occurs in part through regulation of the basolateral K+ channel Kcnj10 (164). Thus, β-adrenergic activation in the DCT may contribute to the blood pressure-raising effects associated with renal sympathetic nerve activation in the kidney. In addition to this, β1-adrenergic receptors are present at low levels throughout the TAL and DCT. Activation of these receptors is well established to regulate renin secretion (155, 165).

Thus, multiple adrenergic receptors expressed in different renal tubule segments are expected to produce a complex aggregate effect on salt and water excretion and blood pressure regulation. Beyond this, adrenergic receptors in renin-secreting juxtaglomerular granular cells, vascular smooth muscle cells, and sympathetic neurons are all likely to make their own contributions to blood pressure regulation (155). The role of renal α-adrenergic receptors in hypertension and kidney function has been reviewed in greater detail by multiple groups (166168).

Lysophosphatidic Acid Receptors

Lysophosphatidic acid receptors (LPARs) are also widely expressed, including Lpar3/6 in the PT and Lpar1 in the CD (Fig. 9). LPA is derived from membrane lipids and is present in most tissues, and its effect is regulated by the specific tissue and cellular distribution of LPARs (169). RNA and protein expression of Lpar1 and Lpar3 has been demonstrated in these segments in prior microdissection studies in rat nephrons (11, 16). Each Lpar is capable of binding to multiple G proteins: LPAR1 can couple to Gαi, Gαq, and Gα12, LPAR3 couples with Gαi and Gαq, and LPAR6 couples with Gαi and Gα12. The precise coupling of each Lpar in the kidney is still currently unknown.

Figure 9.

Figure 9.

Expression of lysophosphatidic acid receptors (LPARs) along the renal tubule. Colors indicate segment boundaries. CD, collecting duct; DCT, distal convoluted tubule; Glom, glomerulus; DTL, descending thin limb; PT, proximal tubule; TAL, thick ascending limb.

LPARs have been shown to play roles in kidney disease models. In models of ischemia-reperfusion injury, blockade of LPAR3 reduced injury, whereas an LPAR3 agonist enhanced kidney injury (24). These results suggested that multiple Lpars with different affinities for LPA have functions in the ischemia-reperfusion injury response. Increased expression of Lpar 1/Lpar3 has also been observed in animal models of diabetic nephropathy (170, 171). In addition, elevated levels of LPA have been correlated with renal dysfunction (170).

To date, these effects have not been linked to a particular LPAR, nephron segment, or cell type. Treatment of microdissected tubules with LPA, in combination with pharmacological inhibitors and engineered in vivo knockout models, would be informative as to the direct effects of LPARs on transport across the nephron and could provide insights to the role of these receptors in disease states. In addition, LPAR-mediated signals interact with other pathways such as EGF or ANG II (172). The effects of LPAR activation in these discrete contexts remains an open question.

Olfactory Receptors Along the Nephron

Olfactory receptors (ORs) are the largest family of GPCRs, with over 1,109 mouse genes represented in our complete GPCR list. In recent years, ORs have been shown to have wide-ranging expression in nonolfactory tissues, where they have functional roles as chemosensors (173175). Here, we show all ORs that have a TPM of 10 or greater at any point along the nephron, yielding 19 ORs (Supplemental Fig. S2). To our knowledge, these most highly expressed ORs have not been characterized in the kidney. In addition, 17 of 19 ORs with a maximum TPM greater than 10 are most highly expressed in DTL1, although the highest expressing OR found in our RNA-seq data set was Olfr373, an orphan receptor almost exclusively expressed in IMCD cells. The vast majority of ORs are still orphans with no known ligand, and creating specific antibodies is challenging due to a high level of homology between ORs (176). Despite this, multiple olfactory receptors have been identified and characterized to have functional roles in the kidney (177181). The functional role of these orphan ORs represents a largely unanswered and potentially important physiological question.

RESOLVING DIFFERENT APPROACHES TO GPCR DETECTION

The expression of well-established receptors (Pth1r, Avpr2, Gcgr, etc.) has largely been confirmed by unbiased omics studies; however, discrepancies are inevitable when comparing results from different methodologies. One notable omission from our data is the expression of any of the dopamine receptors (Drd1–Drd5). Dopamine is a catecholamine that has been studied for decades as a vasodilator and has been shown to have natriuretic effects via interactions with dopamine receptors (182). However, we did not detect expression of any of the dopamine receptors in the renal tubule (9). These receptors have also not been reported in the tubule in other gene expression studies (14, 15). It is unknown why these receptors remain undetected in multiple omics datasets, despite well-established roles for dopamine in the kidney. It is possible that dopamine effects are mediated by adrenergic receptors, as cross talk has been observed in other tissues (183185). In another example, we identified Agtr1a in the PT, but expression of Agtr1a was virtually zero outside of the PT. Nevertheless, multiple studies have identified ANG II actions in the TAL, DCT, and CD, where it has been implicated to play roles in salt and water transport (186190). Although RNA-seq studies typically have a high degree of sensitivity, it is possible that transcript levels for these receptors are below the level of detection in these experiments while simultaneously producing enough protein to mediate receptor function. This could be true if Agtr1a protein had an exceptionally long half-life. As with dopamine, it is also possible that another receptor is responding to the same agonist that is yet to be identified. Resolving these discrepancies will require further functional studies to verify expression of these receptors and what their roles are.

FUNCTIONAL SIGNIFICANCE

Our conclusions are primarily drawn from RNA-seq data, which can provide a more accurate representation of GPCR expression along the renal tubule than can be achieved by proteomic methods due to greater sensitivity. Hypotheses from transcriptomic and proteomic studies ultimately will require follow-up functional studies to learn the true role of the newly identified receptors in the kidney. We therefore cross referenced our findings with other omics datasets including single-cell RNA-seq studies (14, 75), which are useful for identifying GPCR expression patterns in cellular subpopulations. By conducting differential gene expression analysis using kidney cell type-specific markers (12), localization as well as cell type-specific GPCR expression can be confirmed. We have also cross referenced this data with prior proteomics data from microdissected rat kidneys (16). Proteomics approaches to detecting GPCR expression are limited, and a lower level of GPCR expression than proteomic detection thresholds may be sufficient to affect physiological function. In addition, a general lack of specific validated antibodies can make detection with traditional biochemical approaches such as Western blot analysis difficult. Ultimately, neither mRNA nor protein expression necessarily indicate functional activity. Additional factors such as downstream signaling targets are also yet to be uncovered for many of these receptors. For many GPCRs, further experimental validation in animal models is needed to accurately determine receptor function.

CONCLUSIONS

In this review, we provide a new resource for the study of GPCRs in the renal tubule. Using a recently published transcriptome for mouse renal tubule segments, we created a comprehensive list of GPCRs and ORs expressed in the mouse and where they are expressed. This GPCR expression set will help to form new hypotheses for future independent studies to validate GPCR expression and function in cellular and animal models. Given that GPCRs are the most targeted gene group for therapeutics, we hope this will encourage study of lesser known GPCRs that will lead to new discoveries.

SUPPLEMENTAL DATA

Supplemental Figs. S1–S3 and Supplemental Table S1: https://hpcwebapps.cit.nih.gov/ESBL/Database/GPCRSupplemental/.

GRANTS

This work was primarily funded by National Heart, Lung, and Blood Institute Grants ZIAHL001285 and ZIAHL006129 (to M.A.K.). This work was also supported through a National Institutes of Health Bench-to-Bedside award from the National Institutes of Health Office of Clinical Research.

DISCLOSURES

No conflicts of interest, financial or otherwise, are declared by the authors.

AUTHOR CONTRIBUTIONS

B.G.P., L.C., and M.A.K. conceived and designed research; B.G.P., L.C., C-L.C., and M.A.K. analyzed data; B.G.P. and M.A.K. prepared figures; B.G.P., L.C., C-L.C., V.R., and M.A.K. drafted manuscript; B.G.P., L.C., C-L.C., V.R., and M.A.K. edited and revised manuscript; B.G.P., L.C., C-L.C., V.R., and M.A.K. approved final version of manuscript.

APPENDIX

Curation of a Comprehensive List of GPCR Genes in the Mouse, Human, and Rat Genomes

We curated a complete list of mouse GPCRs using multiple online databases. This list includes data from the International Union of Basic and Clinical Pharmacology/British Pharmacological Society (IUPHAR/BPS) (https://www.guidetopharmacology.org/GRAC/GPCRListForward?class=A), Mouse Genome Informatics (MGI; http://www.informatics.jax.org/), and Ensembl Biomart (https://m.ensembl.org/info/data/biomart/index.html). Human and rat orthologs were mapped from data obtained from Ensembl Biomart and the Rat Genome Database (RGD; https://rgd.mcw.edu/). The resulting GPCR lists for the mouse, rat, and human genomes are provided at https://hpcwebapps.cit.nih.gov/ESBL/Database/GPCRs/. This list contains 758 mouse GPCRs and their human and rat orthologs (Fig. 1A).

This list does not include ORs, which we provide as a separate list. For mouse ORs, we obtained a gene list searching for “olfactory receptors” from MGI (http://www.informatics.jax.org/searchtool/Search.do?query=Olfactory+receptor&submit=Quick%0D%0ASearch), yielding 1,109 gene symbols after we filtered out the pseudogenes. We mapped the mouse ORs to human and rat ORs using tools at the Human Genome Organization Gene Nomenclature Committee website (https://www.genenames.org/data/genegroup/#!/group/139) and RGD. This curated list is provided at https://hpcwebapps.cit.nih.gov/ESBL/Database/GPCRs/MouseHumanRatORs.html. We used the comprehensive GPCR and OR lists to extract expression data from our omics datasets, thus providing a complete picture of GPCR expression.

Sorting for GPCRs Selectively Expressed in Renal Tubule Segments

The curated lists of GPCRs and ORs were mapped to RNA-seq data from microdissected renal tubule segments from the mouse generated by Chen et al. (9) (Fig. 1B). This yielded matches to 758 GPCRs and 1,109 ORs across 14 tubule segments as well as the glomerulus. Expression levels for all GPCRs can be found at https://hpcwebapps.cit.nih.gov/ESBL/Database/GPCRs/TubuleTPM.html. GPCRs with a maximum TPM of 10 or less were filtered out to eliminate low-expressing receptors. Only 104 GPCRs and 19 ORs had TPM values of greater than 10 (Fig. 1C). From this, GPCR expression was ranked in each tubule segment (top 10 GPCRs for each segment shown in Table 1). Several top expressed GPCRs were present ubiquitously across all segments, suggesting that these receptors are less responsible for individual segment functions. For this review, we wanted to highlight understudied receptors that have distinct patterns of expression that suggest functional roles. To determine nephron segment-selective expression of GPCRs, we identified the TPM values for each gene in a particular nephron segment group. Groups of nephron segments were combined and analyzed by “regions” as follows: the PT, which includes PTS1, PTS2, and PTS3; DTL, which includes DTL1, DLT2, and DTL3; ATL; MTAL and cortical TAL; DCT; CNT; and CD, which includes the CCD, OMCD, and IMCD. To identify GPCRs selectively expressed in these regions, we compared maximum TPM values for segments within the region with maximum TPM values for segments outside the region (Fig. 1D). A ratio of these two values of 1.5 or greater was considered to be selective for a particular region. These GPCRs are presented based on the segment in which they are selectively expressed (Figs. 2, 3, 4, 5, 6).

Note that glomerular GPCRs were not covered in this review. GPCRs selectively expressed in the glomerulus are provided in Supplemental Fig. S3. Multiple transcriptomic studies have been published recently focusing on the glomerulus, which provide a more complete picture of the glomerular expression landscape (191, 192).

Selectively expressed GPCRs were matched to known ligands, coupled G proteins, and known physiological roles in the corresponding nephron segment. Ligand and G protein coupling data were obtained from the IUPHAR/BPS database (https://www.guidetopharmacology.org) and the GPCRdb database (193) (see Tables 36 and Supplemental Table S1). The curated GPCR list was also mapped to proteomics data obtained from microdissected nephron segments of Sprague–Dawley rats, published in Ref. 16 and available at https://esbl.nhlbi.nih.gov/KTEA/, which yielded 27 GPCRs. The curated GPCR list was also mapped to single-cell RNA-seq data from intercalated and principal cells from mouse CDs, published in Ref. 12 and available at https://hpcwebapps.cit.nih.gov/ESBL/Database/scRNA-Seq/, which yielded 40 GPCRs.

REFERENCES

  • 1.Morel F, Doucet A. Hormonal control of kidney functions at the cell level. Physiol Rev 66: 377–468, 1986. doi: 10.1152/physrev.1986.66.2.377. [DOI] [PubMed] [Google Scholar]
  • 2.Morel F, Imbert-Teboul M, Chabardes D. Distribution of hormone-dependent adenylate cyclase in the nephron and its physiological significance. Annu Rev Physiol 43: 569–581, 1981. doi: 10.1146/annurev.ph.43.030181.003033. [DOI] [PubMed] [Google Scholar]
  • 3.Chabardes D, Imbert M, Clique A, Montegut M, Morel F. PTH sensitive adenyl cyclase activity in different segments of the rabbit nephron. Pflugers Arch 354: 229–239, 1975. doi: 10.1007/BF00584646. [DOI] [PubMed] [Google Scholar]
  • 4.Bailly C, Imbert-Teboul M, Chabardes D, Hus-Citharel A, Montegut M, Clique A, Morel F. The distal nephron of rat kidney: a target site for glucagon. Proc Natl Acad Sci USA 77: 3422–3424, 1980. doi: 10.1073/pnas.77.6.3422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Chabardès D, Imbert-Teboul M, Montégut M, Clique A, Morel F. Distribution of calcitonin-sensitive adenylate cyclase activity along the rabbit kidney tubule. Proc Natl Acad Sci USA 73: 3608–3612, 1976. doi: 10.1073/pnas.73.10.3608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Butlen D, Morel F. Glucagon receptors along the nephron: [125I]glucagon binding in rat tubules. Pflugers Arch 404: 348–353, 1985. doi: 10.1007/BF00585347. [DOI] [PubMed] [Google Scholar]
  • 7.Imbert M, Chabardes D, Montegut M, Clique A, Morel F. Vasopressin dependent adenylate cyclase in single segments of rabbit kidney tubule. Pflugers Arch 357: 173–186, 1975. doi: 10.1007/BF00585973. [DOI] [PubMed] [Google Scholar]
  • 8.Burg MB. Isolated perfused tubule. Introduction: background and development of microperfusion technique. Kidney Int 22: 417–424, 1982. doi: 10.1038/ki.1982.194. [DOI] [PubMed] [Google Scholar]
  • 9.Chen L, Chou C-L, Knepper MA. A comprehensive map of mRNAs and their isoforms across all 14 renal tubule segments of mouse. J Am Soc Nephrol 32: 897–912, 2021. doi: 10.1681/ASN.2020101406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Clark JZ, Chen L, Chou CL, Jung HJ, Lee JW, Knepper MA. Representation and relative abundance of cell-type selective markers in whole-kidney RNA-Seq data. Kidney Int 95: 787–796, 2019. doi: 10.1016/j.kint.2018.11.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Lee JW, Chou CL, Knepper MA. Deep sequencing in microdissected renal tubules identifies nephron segment-specific transcriptomes. J Am Soc Nephrol 26: 2669–2677, 2015. doi: 10.1681/ASN.2014111067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Chen L, Lee JW, Chou C-L, Nair AV, Battistone MA, Păunescu TG, Merkulova M, Breton S, Verlander JW, Wall SM, Brown D, Burg MB, Knepper MA. Transcriptomes of major renal collecting duct cell types in mouse identified by single-cell RNA-seq. Proc Natl Acad Sci USA 114: E9989–E9998, 2017. doi: 10.1073/pnas.1710964114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Park J, Shrestha R, Qiu C, Kondo A, Huang S, Werth M, Li M, Barasch J, Susztak K. Single-cell transcriptomics of the mouse kidney reveals potential cellular targets of kidney disease. Science 360: 758–763, 2018. doi: 10.1126/science.aar2131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Ransick A, Lindstrom NO, Liu J, Zhu Q, Guo JJ, Alvarado GF, Kim AD, Black HG, Kim J, McMahon AP. Single-cell profiling reveals sex, lineage, and regional diversity in the mouse kidney. Dev Cell 51: 399–413.e7, 2019. doi: 10.1016/j.devcel.2019.10.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Wu H, Malone AF, Donnelly EL, Kirita Y, Uchimura K, Ramakrishnan SM, Gaut JP, Humphreys BD. Single-cell transcriptomics of a human kidney allograft biopsy specimen defines a diverse inflammatory response. J Am Soc Nephrol 29: 2069–2080, 2018. doi: 10.1681/ASN.2018020125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Limbutara K, Chou CL, Knepper MA. Quantitative proteomics of all 14 renal tubule segments in rat. J Am Soc Nephrol 31: 1255–1266, 2020. doi: 10.1681/ASN.2020010071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Insel PA, Sriram K, Gorr MW, Wiley SZ, Michkov A, Salmeron C, Chinn AM. GPCRomics: an approach to discover GPCR drug targets. Trends Pharmacol Sci 40: 378–387, 2019. doi: 10.1016/j.tips.2019.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Insel PA, Sriram K, Wiley SZ, Wilderman A, Katakia T, McCann T, Yokouchi H, Zhang L, Corriden R, Liu D, Feigin ME, French RP, Lowy AM, Murray F. GPCRomics: GPCR expression in cancer cells and tumors identifies new, potential biomarkers and therapeutic targets. Front Pharmacol 9: 431, 2018. doi: 10.3389/fphar.2018.00431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Dupré DJ, Robitaille M, Rebois RV, Hébert TE. The role of Gβγ subunits in the organization, assembly, and function of GPCR signaling complexes. Annu Rev Pharmacol Toxicol 49: 31–56, 2009. doi: 10.1146/annurev-pharmtox-061008-103038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Smrcka AV. G protein βγ subunits: central mediators of G protein-coupled receptor signaling. Cell Mol Life Sci 65: 2191–2214, 2008. doi: 10.1007/s00018-008-8006-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Luttrell LM, Lefkowitz RJ. The role of β-arrestins in the termination and transduction of G-protein-coupled receptor signals. J Cell Sci 115: 455–465, 2002. [DOI] [PubMed] [Google Scholar]
  • 22.Feinstein TN, Yui N, Webber MJ, Wehbi VL, Stevenson HP, King JD Jr, Hallows KR, Brown D, Bouley R, Vilardaga J-P. Noncanonical control of vasopressin receptor type 2 signaling by retromer and arrestin. J Biol Chem 288: 27849–27860, 2013. doi: 10.1074/jbc.M112.445098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Vilardaga JP, Gardella TJ, Wehbi VL, Feinstein TN. Non-canonical signaling of the PTH receptor. Trends Pharmacol Sci 33: 423–431, 2012. doi: 10.1016/j.tips.2012.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Okusa MD, Ye H, Huang L, Sigismund L, Macdonald T, Lynch KR. Selective blockade of lysophosphatidic acid LPA3 receptors reduces murine renal ischemia-reperfusion injury. Am J Physiol Renal Physiol 285: F565–F574, 2003. doi: 10.1152/ajprenal.00023.2003. [DOI] [PubMed] [Google Scholar]
  • 25.Takahashi-Iwanaga H, Kimura S, Konno K, Watanabe M, Iwanaga T. Intrarenal signaling mediated by CCK plays a role in salt intake-induced natriuresis. Am J Physiol Renal Physiol 313: F20–F29, 2017. doi: 10.1152/ajprenal.00539.2016. [DOI] [PubMed] [Google Scholar]
  • 26.He W, Miao FJP, Lin DCH, Schwandner RT, Wang Z, Gao J, Chen J-L, Tian H, Ling L. Citric acid cycle intermediates as ligands for orphan G-protein-coupled receptors. Nature 429: 188–193, 2004. doi: 10.1038/nature02488. [DOI] [PubMed] [Google Scholar]
  • 27.Toma I, Kang JJ, Sipos A, Vargas S, Bansal E, Hanner F, Meer E, Peti-Peterdi J. Succinate receptor GPR91 provides a direct link between high glucose levels and renin release in murine and rabbit kidney. J Clin Invest 118: 2526–2534, 2008. doi: 10.1172/JCI33293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.de Gasparo M, Catt KJ, Inagami T, Wright JW, Unger T. International union of pharmacology. XXIII. The angiotensin II receptors. Pharmacol Rev 52: 415–472, 2000. [PubMed] [Google Scholar]
  • 29.Garvin JL. Angiotensin stimulates bicarbonate transport and Na+/K+ ATPase in rat proximal straight tubules. J Am Soc Nephrol 1: 1146–1152, 1991. doi: 10.1681/ASN.V1101146. [DOI] [PubMed] [Google Scholar]
  • 30.Harrison-Bernard LM, Navar LG, Ho MM, Vinson GP, el-Dahr SS. Immunohistochemical localization of ANG II AT1 receptor in adult rat kidney using a monoclonal antibody. Am J Physiol Renal Physiol 273: F170–F177, 1997. doi: 10.1152/ajprenal.1997.273.1.F170. [DOI] [PubMed] [Google Scholar]
  • 31.Liu Y, Rafferty TM, Rhee SW, Webber JS, Song L, Ko B, Hoover RS, He B, Mu S. CD8+ T cells stimulate Na-Cl co-transporter NCC in distal convoluted tubules leading to salt-sensitive hypertension. Nat Commun 8: 14037, 2017. doi: 10.1038/ncomms14037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Beach RE, Schwab SJ, Brazy PC, Dennis VW. Norepinephrine increases Na+/K+-ATPase and solute transport in rabbit proximal tubules. Am J Physiol Renal Physiol 252: F215–F220, 1987. doi: 10.1152/ajprenal.1987.252.2.F215. [DOI] [PubMed] [Google Scholar]
  • 33.Bylund DB, Ray-Prenger C. α-2A and α-2B adrenergic receptor subtypes: attenuation of cyclic AMP production in cell lines containing only one receptor subtype. J Pharmacol Exp Ther 251: 640–644, 1989. [PubMed] [Google Scholar]
  • 34.Cussac D, Schaak S, Gales C, Flordellis C, Denis C, Paris H. α(2B)-Adrenergic receptors activate MAPK and modulate proliferation of primary cultured proximal tubule cells. Am J Physiol Renal Physiol 282: F943–F952, 2002. doi: 10.1152/ajprenal.0108.2001. [DOI] [PubMed] [Google Scholar]
  • 35.Hague C, Chen Z, Uberti M, Minneman KP. α(1)-Adrenergic receptor subtypes: non-identical triplets with different dancing partners? Life Sci 74: 411–418, 2003. doi: 10.1016/j.lfs.2003.07.008. [DOI] [PubMed] [Google Scholar]
  • 36.Liu F, Nesbitt T, Drezner MK, Friedman PA, Gesek FA. Proximal nephron Na+/H+ exchange is regulated by α1A- and α1B- and α 1B-adrenergic receptor subtypes. Mol Pharmacol 52: 1010–1018, 1997. doi: 10.1124/mol.52.6.1010. [DOI] [PubMed] [Google Scholar]
  • 37.Amizuka N, Lee HS, Kwan MY, Arazani A, Warshawsky H, Hendy GN, Ozawa H, White JH, Goltzman D. Cell-specific expression of the parathyroid hormone (PTH)/PTH-related peptide receptor gene in kidney from kidney-specific and ubiquitous promoters. Endocrinology 138: 469–481, 1997. doi: 10.1210/endo.138.1.4845. [DOI] [PubMed] [Google Scholar]
  • 38.Dennis VW. Influence of bicarbonate on parathyroid hormone-induced changes in fluid absorption by the proximal tubule. Kidney Int 10: 373–380, 1976. doi: 10.1038/ki.1976.123. [DOI] [PubMed] [Google Scholar]
  • 39.Plati J, Tsomaia N, Piserchio A, Mierke DF. Structural features of parathyroid hormone receptor coupled to Gα(s)-protein. Biophys J 92: 535–540, 2007. doi: 10.1529/biophysj.106.094813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Ba J, Brown D, Friedman PA. Calcium-sensing receptor regulation of PTH-inhibitable proximal tubule phosphate transport. Am J Physiol Renal Physiol 285: F1233–F1243, 2003. doi: 10.1152/ajprenal.00249.2003. [DOI] [PubMed] [Google Scholar]
  • 41.Traebert M, Volkl H, Biber J, Murer H, Kaissling B. Luminal and contraluminal action of 1–34 and 3–34 PTH peptides on renal type IIa Na-Pi cotransporter. Am J Physiol Renal Physiol 278: F792–F798, 2000. doi: 10.1152/ajprenal.2000.278.5.F792. [DOI] [PubMed] [Google Scholar]
  • 42.Dennis VW, Bello-Reuss E, Robinson RR. Response of phosphate transport to parathyroid hormone in segments of rabbit nephron. Am J Physiol Renal Physiol 233: F29–F38, 1977. doi: 10.1152/ajprenal.1977.233.1.F29. [DOI] [PubMed] [Google Scholar]
  • 43.Yanagawa N, Jo OD. Possible role of calcium in parathyroid hormone actions in rabbit renal proximal tubules. Am J Physiol Renal Physiol 250: F942–F948, 1986. doi: 10.1152/ajprenal.1986.250.5.F942. [DOI] [PubMed] [Google Scholar]
  • 44.Gurley SB, Riquier-Brison ADM, Schnermann J, Sparks MA, Allen AM, Haase VH, Snouwaert JN, Le TH, McDonough AA, Koller BH, Coffman TM. AT1A angiotensin receptors in the renal proximal tubule regulate blood pressure. Cell Metab 13: 469–475, 2011. doi: 10.1016/j.cmet.2011.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Li H, Weatherford ET, Davis DR, Keen HL, Grobe JL, Daugherty A, Cassis LA, Allen AM, Sigmund CD. Renal proximal tubule angiotensin AT1A receptors regulate blood pressure. Am J Physiol Regul Integr Comp Physiol 301: R1067–R1077, 2011. doi: 10.1152/ajpregu.00124.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Liu FY, Cogan MG. Angiotensin II stimulates early proximal bicarbonate absorption in the rat by decreasing cyclic adenosine monophosphate. J Clin Invest 84: 83–91, 1989. doi: 10.1172/JCI114174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Poggioli J, Lazar G, Houillier P, Gardin JP, Achard JM, Paillard M. Effects of angiotensin II and nonpeptide receptor antagonists on transduction pathways in rat proximal tubule. Am J Physiol Cell Physiol 263: C750–C758, 1992. doi: 10.1152/ajpcell.1992.263.4.C750. [DOI] [PubMed] [Google Scholar]
  • 48.Gill JR Jr, Casper AG. Effect of renal α-adrenergic stimulation on proximal tubular sodium reabsorption. Am J Physiol 223: 1201–1205, 1972. doi: 10.1152/ajplegacy.1972.223.5.1201. [DOI] [PubMed] [Google Scholar]
  • 49.Jackson CA, Insel PA. Renal α-adrenergic receptors and genetic hypertension. Pediatr Nephrol 7: 853–858, 1993. doi: 10.1007/BF01213373. [DOI] [PubMed] [Google Scholar]
  • 50.Umemura S, Marver D, Smyth DD, Pettinger WA. α2-Adrenoceptors and cellular cAMP levels in single nephron segments from the rat. Am J Physiol Renal Physiol 249: F28–F33, 1985. doi: 10.1152/ajprenal.1985.249.1.F28. [DOI] [PubMed] [Google Scholar]
  • 51.Pfeil EM, Brands J, Merten N, Vogtle T, Vescovo M, Rick U, Albrecht IM, Heycke N, Kawakami K, Ono Y, Ngako Kadji FM, Hiratsuka S, Aoki J, Haberlein F, Matthey M, Garg J, Hennen S, Jobin ML, Seier K, Calebiro D, Pfeifer A, Heinemann A, Wenzel D, Konig GM, Nieswandt B, Fleischmann BK, Inoue A, Simon K, Kostenis E. Heterotrimeric G protein subunit gαq is a master switch for gβγ-mediated calcium mobilization by Gi-coupled GPCRs. Mol Cell 80: 940–954.e6, 2020. doi: 10.1016/j.molcel.2020.10.027. [DOI] [PubMed] [Google Scholar]
  • 52.Sadagopan N, Li W, Roberds SL, Major T, Preston GM, Yu Y, Tones MA. Circulating succinate is elevated in rodent models of hypertension and metabolic disease. Am J Hypertens 20: 1209–1215, 2007. doi: 10.1016/j.amjhyper.2007.05.010. [DOI] [PubMed] [Google Scholar]
  • 53.Correa PR, Kruglov EA, Thompson M, Leite MF, Dranoff JA, Nathanson MH. Succinate is a paracrine signal for liver damage. J Hepatol 47: 262–269, 2007. doi: 10.1016/j.jhep.2007.03.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Dufresne M, Seva C, Fourmy D. Cholecystokinin and gastrin receptors. Physiol Rev 86: 805–847, 2006. doi: 10.1152/physrev.00014.2005. [DOI] [PubMed] [Google Scholar]
  • 55.Lacourse KA, Lay JM, Swanberg LJ, Jenkins C, Samuelson LC. Molecular structure of the mouse CCK-A receptor gene. Biochem Biophys Res Commun 236: 630–635, 1997. doi: 10.1006/bbrc.1997.7030. [DOI] [PubMed] [Google Scholar]
  • 56.Monstein HJ, Nylander AG, Salehi A, Chen D, Lundquist I, Hakanson R. Cholecystokinin-A and cholecystokinin-B/gastrin receptor mRNA expression in the gastrointestinal tract and pancreas of the rat and man. A polymerase chain reaction study. Scand J Gastroenterol 31: 383–390, 1996. doi: 10.3109/00365529609006415. [DOI] [PubMed] [Google Scholar]
  • 57.Veiras LC, Girardi ACC, Curry J, Pei L, Ralph DL, Tran A, Castelo-Branco RC, Pastor-Soler N, Arranz CT, Yu ASL, McDonough AA. Sexual dimorphic pattern of renal transporters and electrolyte homeostasis. J Am Soc Nephrol 28: 3504–3517, 2017. doi: 10.1681/ASN.2017030295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Breyer MD, Jacobson HR, Davis LS, Breyer RM. In situ hybridization and localization of mRNA for the rabbit prostaglandin EP3 receptor. Kidney Int 44: 1372–1378, 1993. doi: 10.1038/ki.1993.391. [DOI] [PubMed] [Google Scholar]
  • 59.Gu R, Jin Y, Zhai Y, Yang L, Zhang C, Li W, Wang L, Kong S, Zhang Y, Yang B, Wang WH. PGE2 inhibits basolateral 50 pS potassium channels in the thick ascending limb of the rat kidney. Kidney Int 74: 478–485, 2008. doi: 10.1038/ki.2008.198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Caride AJ, Chini EN, Homma S, Penniston JT, Dousa TP. mRNA encoding four isoforms of the plasma membrane calcium pump and their variants in rat kidney and nephron segments. J Lab Clin Med 132: 149–156, 1998. doi: 10.1016/S0022-2143(98)90010-5. [DOI] [PubMed] [Google Scholar]
  • 61.Motoyama HI, Friedman PA. Calcium-sensing receptor regulation of PTH-dependent calcium absorption by mouse cortical ascending limbs. Am J Physiol Renal Physiol 283: F399–F406, 2002. doi: 10.1152/ajprenal.00346.2001. [DOI] [PubMed] [Google Scholar]
  • 62.Bankir L, Bouby N, Blondeau B, Crambert G. Glucagon actions on the kidney revisited: possible role in potassium homeostasis. Am J Physiol Renal Physiol 311: F469–F486, 2016. doi: 10.1152/ajprenal.00560.2015. [DOI] [PubMed] [Google Scholar]
  • 63.Riccardi D, Lee WS, Lee K, Segre GV, Brown EM, Hebert SC. Localization of the extracellular Ca2+-sensing receptor and PTH/PTHrP receptor in rat kidney. Am J Physiol Renal Physiol 271: F951–F956, 1996. doi: 10.1152/ajprenal.1996.271.4.F951. [DOI] [PubMed] [Google Scholar]
  • 64.Morel F, Butlen D. [17] Hormonal receptors in the isolated tubule. In: Biomembranes Part V: Cellular and Subcellular Transport: Epithelial Cells. Academic Press, 1990, p. 303–325. [DOI] [PubMed] [Google Scholar]
  • 65.Di Stefano A, Wittner M, Nitschke R, Braitsch R, Greger R, Bailly C, Amiel C, Roinel N, de Rouffignac C. Effects of parathyroid hormone and calcitonin on Na+, Cl, K+, Mg2+, and Ca2+ transport in cortical and medullary thick ascending limbs of mouse kidney. Pflugers Arch 417: 161–167, 1990. doi: 10.1007/BF00370694. [DOI] [PubMed] [Google Scholar]
  • 66.Hall DA, Varney DM. Effect of vasopressin on electrical potential difference and chloride transport in mouse medullary thick ascending limb of Henle’s loop. J Clin Invest 66: 792–802, 1980. doi: 10.1172/JCI109917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Hebert SC, Culpepper RM, Andreoli TE. NaCl transport in mouse medullary thick ascending limbs. II. ADH enhancement of transcellular NaCl cotransport; origin of transepithelial voltage. Am J Physiol Renal Physiol 241: F432–F442, 1981. doi: 10.1152/ajprenal.1981.241.4.F432. [DOI] [PubMed] [Google Scholar]
  • 68.Sasaki S, Imai M. Effects of vasopressin on water and NaCl transport across the in vitro perfused medullary thick ascending limb of Henle's loop of mouse, rat, and rabbit kidneys. Pflugers Arch 383: 215–221, 1980. doi: 10.1007/BF00587521. [DOI] [PubMed] [Google Scholar]
  • 69.Wittner M, Di Stefano A. Effects of antidiuretic hormone, parathyroid hormone and glucagon on transepithelial voltage and resistance of the cortical and medullary thick ascending limb of Henle's loop of the mouse nephron. Pflugers Arch 415: 707–712, 1990. doi: 10.1007/BF02584009. [DOI] [PubMed] [Google Scholar]
  • 70.Stokes JB. Effect of prostaglandin E2 on chloride transport across the rabbit thick ascending limb of Henle. Selective inhibitions of the medullary portion. J Clin Invest 64: 495–502, 1979. doi: 10.1172/JCI109487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Olinski LE, Lin EM, Oancea E. Illuminating insights into opsin 3 function in the skin. Adv Biol Regul 75: 100668, 2020. doi: 10.1016/j.jbior.2019.100668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Olinski LE, Tsuda AC, Kauer JA, Oancea E. Endogenous opsin 3 (OPN3) protein expression in the adult brain using a novel OPN3-mCherry knock-in mouse model. ENEURO 7: 0107–0120, 2020. doi: 10.1523/ENEURO.0107-20.2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Sato M, Tsuji T, Yang K, Ren X, Dreyfuss JM, Huang TL, Wang CH, Shamsi F, Leiria LO, Lynes MD, Yau KW, Tseng YH. Cell-autonomous light sensitivity via Opsin3 regulates fuel utilization in brown adipocytes. PLoS Biol 18: e3000630, 2020. doi: 10.1371/journal.pbio.3000630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Briggs JP, Lorenz JN, Weihprecht H, Schnermann J. Macula densa control of renin secretion. Ren Physiol Biochem 14: 164–174, 1991. doi: 10.1159/000173402. [DOI] [PubMed] [Google Scholar]
  • 75.Chen L, Chou C-L, Knepper MA. Targeted single-cell RNA-seq identifies minority cell types of kidney distal nephron. J Am Soc Nephrol 32: 886–896, 2021. doi: 10.1681/ASN.2020101407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Aoyagi T, Izumi Y, Hiroyama M, Matsuzaki T, Yasuoka Y, Sanbe A, Miyazaki H, Fujiwara Y, Nakayama Y, Kohda Y, Yamauchi J, Inoue T, Kawahara K, Saito H, Tomita K, Nonoguchi H, Tanoue A. Vasopressin regulates the renin-angiotensin-aldosterone system via V1a receptors in macula densa cells. Am J Physiol Renal Physiol 295: F100–F107, 2008. doi: 10.1152/ajprenal.00088.2008. [DOI] [PubMed] [Google Scholar]
  • 77.Davies T, Marians R, Latif R. The TSH receptor reveals itself. J Clin Invest 110: 161–164, 2002. doi: 10.1172/JCI16234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Battistone MA, Nair AV, Barton CR, Liberman RN, Peralta MA, Capen DE, Brown D, Breton S. extracellular adenosine stimulates vacuolar ATPase-dependent proton secretion in medullary intercalated cells. J Am Soc Nephrol 29: 545–556, 2018. doi: 10.1681/ASN.2017060643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Vitzthum H, Weiss B, Bachleitner W, Kramer BK, Kurtz A. Gene expression of adenosine receptors along the nephron. Kidney Int 65: 1180–1190, 2004. doi: 10.1111/j.1523-1755.2004.00490.x. [DOI] [PubMed] [Google Scholar]
  • 80.Zhang W, Zhang Y, Wang W, Dai Y, Ning C, Luo R, Sun K, Glover L, Grenz A, Sun H, Tao L, Zhang W, Colgan SP, Blackburn MR, Eltzschig HK, Kellems RE, Xia Y. Elevated ecto-5'-nucleotidase-mediated increased renal adenosine signaling via A2B adenosine receptor contributes to chronic hypertension. Circ Res 112: 1466–1478, 2013. doi: 10.1161/CIRCRESAHA.111.300166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Saito O, Guan Y, Qi Z, Davis LS, Komhoff M, Sugimoto Y, Narumiya S, Breyer RM, Breyer MD. Expression of the prostaglandin F receptor (FP) gene along the mouse genitourinary tract. Am J Physiol Renal Physiol 284: F1164–F1170, 2003. doi: 10.1152/ajprenal.00441.2002. [DOI] [PubMed] [Google Scholar]
  • 82.Wang L, Zhang C, Su XT, Lin DH, Wu P, Schwartzman ML, Wang WH. PGF2α regulates the basolateral K channels in the distal convoluted tubule. Am J Physiol Renal Physiol 313: F254–F261, 2017. doi: 10.1152/ajprenal.00102.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Wang LJ, Xiao Y, Fang J, Wang JL, Zhang H, Meng XX, Gong RL, Gu R. PGF2α stimulates the 10-pS Cl channel and thiazide-sensitive Na+-Cl cotransporter in the distal convoluted tubule. Am J Physiol Renal Physiol 319: F414–F422, 2020. doi: 10.1152/ajprenal.00287.2020. [DOI] [PubMed] [Google Scholar]
  • 84.Berg P, Svendsen SL, Sorensen MV, Larsen CK, Andersen JF, Jensen-Fangel S, Jeppesen M, Schreiber R, Cabrita I, Kunzelmann K, Leipziger J. Impaired renal HCO3 excretion in cystic fibrosis. J Am Soc Nephrol 31: 1711–1727, 2020. doi: 10.1681/ASN.2020010053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Chu JY, Chung SC, Lam AK, Tam S, Chung SK, Chow BK. Phenotypes developed in secretin receptor-null mice indicated a role for secretin in regulating renal water reabsorption. Mol Cell Biol 27: 2499–2511, 2007. doi: 10.1128/MCB.01088-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Lazo-Fernandez Y, Welling PA, Wall SM. α-Ketoglutarate stimulates pendrin-dependent Cl absorption in the mouse CCD through protein kinase C. Am J Physiol Renal Physiol 315: F7–F15, 2018. doi: 10.1152/ajprenal.00576.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Moriyama T, Garcia-Perez A, Burg MB. Osmotic regulation of aldose reductase protein synthesis in renal medullary cells. J Biol Chem 264: 16810–16814, 1989. [PubMed] [Google Scholar]
  • 88.Carrasco MP, Asboth G, Phaneuf S, Lopez Bernal A. Activation of the prostaglandin FP receptor in human granulosa cells. J Reprod Fertil 111: 309–317, 1997. doi: 10.1530/jrf.0.1110309. [DOI] [PubMed] [Google Scholar]
  • 89.Ito S, Sakamoto K, Mochizuki-Oda N, Ezashi T, Miwa K, Okuda-Ashitaka E, Shevchenko VI, Kiso Y, Hayaishi O. Prostaglandin F2 α receptor is coupled to Gq in cDNA-transfected Chinese hamster ovary cells. Biochem Biophys Res Commun 200: 756–762, 1994. doi: 10.1006/bbrc.1994.1515. [DOI] [PubMed] [Google Scholar]
  • 90.Viteri AL, Poppell JW, Lasater JM, Dyck WP. Renal response to secretin. J Appl Physiol 38: 661–664, 1975. doi: 10.1152/jappl.1975.38.4.661. [DOI] [PubMed] [Google Scholar]
  • 91.Procino G, Milano S, Carmosino M, Barbieri C, Nicoletti MC, Li JH, Wess J, Svelto M. Combination of secretin and fluvastatin ameliorates the polyuria associated with X-linked nephrogenic diabetes insipidus in mice. Kidney Int 86: 127–138, 2014. doi: 10.1038/ki.2014.10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Tokonami N, Morla L, Centeno G, Mordasini D, Ramakrishnan SK, Nikolaeva S, Wagner CA, Bonny O, Houillier P, Doucet A, Firsov D. α-Ketoglutarate regulates acid-base balance through an intrarenal paracrine mechanism. J Clin Invest 123: 3166–3171, 2013. doi: 10.1172/JCI67562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Dijksterhuis JP, Petersen J, Schulte G. WNT/Frizzled signalling: receptor-ligand selectivity with focus on FZD-G protein signalling and its physiological relevance: IUPHAR Review 3. Br J Pharmacol 171: 1195–1209, 2014. doi: 10.1111/bph.12364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Itaranta P, Lin Y, Perasaari J, Roel G, Destree O, Vainio S. Wnt-6 is expressed in the ureter bud and induces kidney tubule development in vitro. Genesis 32: 259–268, 2002. doi: 10.1002/gene.10079. [DOI] [PubMed] [Google Scholar]
  • 95.Majumdar A, Vainio S, Kispert A, McMahon J, McMahon AP. Wnt11 and Ret/Gdnf pathways cooperate in regulating ureteric branching during metanephric kidney development. Development 130: 3175–3185, 2003. doi: 10.1242/dev.00520. [DOI] [PubMed] [Google Scholar]
  • 96.Maretto S, Cordenonsi M, Dupont S, Braghetta P, Broccoli V, Hassan AB, Volpin D, Bressan GM, Piccolo S. Mapping Wnt/β-catenin signaling during mouse development and in colorectal tumors. Proc Natl Acad Sci USA 100: 3299–3304, 2003. doi: 10.1073/pnas.0434590100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Chen DQ, Cao G, Chen H, Liu D, Su W, Yu XY, Vaziri ND, Liu XH, Bai X, Zhang L, Zhao YY. Gene and protein expressions and metabolomics exhibit activated redox signaling and wnt/β-catenin pathway are associated with metabolite dysfunction in patients with chronic kidney disease. Redox Biol 12: 505–521, 2017. doi: 10.1016/j.redox.2017.03.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.He W, Kang YS, Dai C, Liu Y. Blockade of Wnt/β-catenin signaling by paricalcitol ameliorates proteinuria and kidney injury. J Am Soc Nephrol 22: 90–103, 2011. doi: 10.1681/ASN.2009121236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Zhou T, He X, Cheng R, Zhang B, Zhang RR, Chen Y, Takahashi Y, Murray AR, Lee K, Gao G, Ma J-X. Implication of dysregulation of the canonical wingless-type MMTV integration site (WNT) pathway in diabetic nephropathy. Diabetologia 55: 255–266, 2012. doi: 10.1007/s00125-011-2314-2. [DOI] [PubMed] [Google Scholar]
  • 100.Pode-Shakked N, Pleniceanu O, Gershon R, Shukrun R, Kanter I, Bucris E, Pode-Shakked B, Tam G, Tam H, Caspi R, Pri-Chen S, Vax E, Katz G, Omer D, Harari-Steinberg O, Kalisky T, Dekel B. Dissecting stages of human kidney development and tumorigenesis with surface markers affords simple prospective purification of nephron stem cells. Sci Rep 6: 23562, 2016. doi: 10.1038/srep23562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Guan Y, Zhang Y, Breyer RM, Fowler B, Davis L, Hebert RL, Breyer MD. Prostaglandin E2 inhibits renal collecting duct Na+ absorption by activating the EP1 receptor. J Clin Invest 102: 194–201, 1998. doi: 10.1172/JCI2872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Hebert RL, Jacobson HR, Breyer MD. PGE2 inhibits AVP-induced water flow in cortical collecting ducts by protein kinase C activation. Am J Physiol Renal Physiol 259: F318–F325, 1990. doi: 10.1152/ajprenal.1990.259.2.F318. [DOI] [PubMed] [Google Scholar]
  • 103.Azroyan A, Cortez-Retamozo V, Bouley R, Liberman R, Ruan YC, Kiselev E, Jacobson KA, Pittet MJ, Brown D, Breton S. Renal intercalated cells sense and mediate inflammation via the P2Y14 receptor. PLoS One 10: e0121419, 2015. doi: 10.1371/journal.pone.0121419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Battistone MA, Mendelsohn AC, Spallanzani RG, Allegretti AS, Liberman RN, Sesma J, Kalim S, Wall SM, Bonventre JV, Lazarowski ER, Brown D, Breton S. Proinflammatory P2Y14 receptor inhibition protects against ischemic acute kidney injury in mice. J Clin Invest 130: 3734–3749, 2020. doi: 10.1172/JCI134791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Zaidman NA, Tomilin VN, Hassanzadeh Khayyat N, Damarla M, Tidmore J, Capen DE, Brown D, Pochynyuk OM, Pluznick JL. Adhesion-GPCR Gpr116 (ADGRF5) expression inhibits renal acid secretion. Proc Natl Acad Sci USA 117: 26470–26481, 2020. doi: 10.1073/pnas.2007620117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Oberhauser V, Vonend O, Rump LC. Neuropeptide Y and ATP interact to control renovascular resistance in the rat. J Am Soc Nephrol 10: 1179–1185, 1999. doi: 10.1681/ASN.V1061179. [DOI] [PubMed] [Google Scholar]
  • 107.Birnbaumer M. Vasopressin receptors. Trends Endocrinol Metab 11: 406–410, 2000. doi: 10.1016/s1043-2760(00)00304-0. [DOI] [PubMed] [Google Scholar]
  • 108.Grantham JJ, Orloff J. Effect of prostaglandin E1 on the permeability response of the isolated collecting tubule to vasopressin, adenosine 3',5'-monophosphate, and theophylline. J Clin Invest 47: 1154–1161, 1968. doi: 10.1172/JCI105804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Hofmeister MV, Damkier HH, Christensen BM, Olde B, Fredrik Leeb-Lundberg LM, Fenton RA, Praetorius HA, Praetorius J. 17β-Estradiol induces nongenomic effects in renal intercalated cells through G protein-coupled estrogen receptor 1. Am J Physiol Renal Physiol 302: F358–F368, 2012. doi: 10.1152/ajprenal.00343.2011. [DOI] [PubMed] [Google Scholar]
  • 110.Bajwa A, Huang L, Ye H, Dondeti K, Song S, Rosin DL, Lynch KR, Lobo PI, Li L, Okusa MD. Dendritic cell sphingosine 1-phosphate receptor-3 regulates Th1-Th2 polarity in kidney ischemia-reperfusion injury. J Immunol 189: 2584–2596, 2012. doi: 10.4049/jimmunol.1200999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Blount MA, Klein JD, Martin CF, Tchapyjnikov D, Sands JM. Forskolin stimulates phosphorylation and membrane accumulation of UT-A3. Am J Physiol Renal Physiol 293: F1308–F1313, 2007. doi: 10.1152/ajprenal.00197.2007. [DOI] [PubMed] [Google Scholar]
  • 112.Sands JM, Nonoguchi H, Knepper MA. Vasopressin effects on urea and H2O transport in inner medullary collecting duct subsegments. Am J Physiol Renal Physiol 253: F823–F832, 1987. doi: 10.1152/ajprenal.1987.253.5.F823. [DOI] [PubMed] [Google Scholar]
  • 113.Zhang C, Sands JM, Klein JD. Vasopressin rapidly increases phosphorylation of UT-A1 urea transporter in rat IMCDs through PKA. Am J Physiol Renal Physiol 282: F85–F90, 2002. doi: 10.1152/ajprenal.0054.2001. [DOI] [PubMed] [Google Scholar]
  • 114.Reif MC, Troutman SL, Schafer JA. Sodium transport by rat cortical collecting tubule. Effects of vasopressin and desoxycorticosterone. J Clin Invest 77: 1291–1298, 1986. doi: 10.1172/JCI112433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Tomita K, Pisano JJ, Knepper MA. Control of sodium and potassium transport in the cortical collecting duct of the rat. Effects of bradykinin, vasopressin, and deoxycorticosterone. J Clin Invest 76: 132–136, 1985. doi: 10.1172/JCI111935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Brooks HL, Ageloff S, Kwon TH, Brandt W, Terris JM, Seth A, Michea L, Nielsen S, Fenton R, Knepper MA. cDNA array identification of genes regulated in rat renal medulla in response to vasopressin infusion. Am J Physiol Renal Physiol 284: F218–F228, 2003. doi: 10.1152/ajprenal.00054.2002. [DOI] [PubMed] [Google Scholar]
  • 117.Ecelbarger CA, Kim GH, Terris J, Masilamani S, Mitchell C, Reyes I, Verbalis JG, Knepper MA. Vasopressin-mediated regulation of epithelial sodium channel abundance in rat kidney. Am J Physiol Renal Physiol 279: F46–F53, 2000. doi: 10.1152/ajprenal.2000.279.1.F46. [DOI] [PubMed] [Google Scholar]
  • 118.Nicco C, Wittner M, DiStefano A, Jounier S, Bankir L, Bouby N. Chronic exposure to vasopressin upregulates ENaC and sodium transport in the rat renal collecting duct and lung. Hypertension 38: 1143–1149, 2001. doi: 10.1161/hy1001.092641. [DOI] [PubMed] [Google Scholar]
  • 119.Perlewitz A, Nafz B, Skalweit A, Fahling M, Persson PB, Thiele BJ. Aldosterone and vasopressin affect α- and γ-ENaC mRNA translation. Nucleic Acids Res 38: 5746–5760, 2010. doi: 10.1093/nar/gkq267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Chen L, Clark JZ, Nelson JW, Kaissling B, Ellison DH, Knepper MA. Renal-tubule epithelial cell nomenclature for single-cell RNA-sequencing studies. J Am Soc Nephrol 30: 1358–1364, 2019. [Erratum in J Am Soc Nephrol 30: 2475, 2019]. doi: 10.1681/ASN.2019040415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Gao Y, Wang HY. Casein kinase 2 is activated and essential for Wnt/β-catenin signaling. J Biol Chem 281: 18394–18400, 2006. doi: 10.1074/jbc.M601112200. [DOI] [PubMed] [Google Scholar]
  • 122.Gao Y, Wang HY. Inositol pentakisphosphate mediates Wnt/β-catenin signaling. J Biol Chem 282: 26490–26502, 2007. doi: 10.1074/jbc.M702106200. [DOI] [PubMed] [Google Scholar]
  • 123.Fricks IP, Carter RL, Lazarowski ER, Harden TK. Gi-dependent cell signaling responses of the human P2Y14 receptor in model cell systems. J Pharmacol Exp Ther 330: 162–168, 2009. doi: 10.1124/jpet.109.150730. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Matsumoto M, Nomura T, Momose K, Ikeda Y, Kondou Y, Akiho H, Togami J, Kimura Y, Okada M, Yamaguchi T. Inactivation of a novel neuropeptide Y/peptide YY receptor gene in primate species. J Biol Chem 271: 27217–27220, 1996. doi: 10.1074/jbc.271.44.27217. [DOI] [PubMed] [Google Scholar]
  • 125.Larhammar D, Blomqvist AG, Yee F, Jazin E, Yoo H, Wahlested C. Cloning and functional expression of a human neuropeptide Y/peptide YY receptor of the Y1 type. J Biol Chem 267: 10935–10938, 1992. [PubMed] [Google Scholar]
  • 126.Lundell I, Blomqvist AG, Berglund MM, Schober DA, Johnson D, Statnick MA, Gadski RA, Gehlert DR, Larhammar D. Cloning of a human receptor of the NPY receptor family with high affinity for pancreatic polypeptide and peptide YY. J Biol Chem 270: 29123–29128, 1995. doi: 10.1074/jbc.270.49.29123. [DOI] [PubMed] [Google Scholar]
  • 127.Adrian TE, Ferri GL, Bacarese-Hamilton AJ, Fuessl HS, Polak JM, Bloom SR. Human distribution and release of a putative new gut hormone, peptide YY. Gastroenterology 89: 1070–1077, 1985. doi: 10.1016/0016-5085(85)90211-2. [DOI] [PubMed] [Google Scholar]
  • 128.Hazelwood RL. The pancreatic polypeptide (PP-Fold) family: gastrointestinal, vascular, and feeding behavioral implications. Proc Soc Exp Biol Med 202: 44–63, 1993. doi: 10.3181/00379727-202-43511g. [DOI] [PubMed] [Google Scholar]
  • 129.Grouzmann E, Alvarez-Bolado G, Meyer C, Osterheld MC, Burnier M, Brunner HR, Waeber B. Localization of neuropeptide Y and its C-terminal flanking peptide in human renal tissue. Peptides 15: 1377–1382, 1994. doi: 10.1016/0196-9781(94)90112-0. [DOI] [PubMed] [Google Scholar]
  • 130.Dillingham MA, Anderson RJ. Mechanism of neuropeptide Y inhibition of vasopressin action in rat cortical collecting tubule. Am J Physiol Renal Physiol 256: F408–F413, 1989. doi: 10.1152/ajprenal.1989.256.3.F408. [DOI] [PubMed] [Google Scholar]
  • 131.Winaver J, Abassi Z. Role of neuropeptide Y in the regulation of kidney function. In: NPY Family of Peptides in Neurobiology, Cardiovascular and Metabolic Disorders: from Genes to Therapeutics, edited by Zukowska Z, Feuerstein GZ.. Basel: Birkhäuser Basel, 2006, p. 123–132. [Google Scholar]
  • 132.Filardo EJ, Quinn JA, Bland KI, Frackelton AR Jr.. Estrogen-induced activation of Erk-1 and Erk-2 requires the G protein-coupled receptor homolog, GPR30, and occurs via trans-activation of the epidermal growth factor receptor through release of HB-EGF. Mol Endocrinol 14: 1649–1660, 2000. doi: 10.1210/mend.14.10.0532. [DOI] [PubMed] [Google Scholar]
  • 133.O'Dowd BF, Nguyen T, Marchese A, Cheng R, Lynch KR, Heng HH, Kolakowski LF Jr, George SR. Discovery of three novel G-protein-coupled receptor genes. Genomics 47: 310–313, 1998. doi: 10.1006/geno.1998.5095. [DOI] [PubMed] [Google Scholar]
  • 134.Revankar CM, Cimino DF, Sklar LA, Arterburn JB, Prossnitz ER. A transmembrane intracellular estrogen receptor mediates rapid cell signaling. Science 307: 1625–1630, 2005. doi: 10.1126/science.1106943. [DOI] [PubMed] [Google Scholar]
  • 135.Grimont A, Bloch-Faure M, El Abida B, Crambert G. Mapping of sex hormone receptors and their modulators along the nephron of male and female mice. FEBS Lett 583: 1644–1648, 2009. doi: 10.1016/j.febslet.2009.04.032. [DOI] [PubMed] [Google Scholar]
  • 136.Cheng L, Poulsen SB, Wu Q, Esteva-Font C, Olesen ETB, Peng L, Olde B, Leeb-Lundberg LMF, Pisitkun T, Rieg T, Dimke H, Fenton RA. Rapid aldosterone-mediated signaling in the DCT increases activity of the thiazide-sensitive NaCl cotransporter. J Am Soc Nephrol 30: 1454–1470, 2019. doi: 10.1681/ASN.2018101025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Gohar EY. G protein-coupled estrogen receptor 1 as a novel regulator of blood pressure. Am J Physiol Renal Physiol 319: F612–F617, 2020. doi: 10.1152/ajprenal.00045.2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Maeda Y, Matsuyuki H, Shimano K, Kataoka H, Sugahara K, Chiba K. Migration of CD4 T cells and dendritic cells toward sphingosine 1-phosphate (S1P) is mediated by different receptor subtypes: S1P regulates the functions of murine mature dendritic cells via S1P receptor type 3. J Immunol 178: 3437–3446, 2007. doi: 10.4049/jimmunol.178.6.3437. [DOI] [PubMed] [Google Scholar]
  • 139.Ancellin N, Hla T. Differential pharmacological properties and signal transduction of the sphingosine 1-phosphate receptors EDG-1, EDG-3, and EDG-5. J Biol Chem 274: 18997–19002, 1999. doi: 10.1074/jbc.274.27.18997. [DOI] [PubMed] [Google Scholar]
  • 140.Lee JW, Huang BX, Kwon H, Rashid MA, Kharebava G, Desai A, Patnaik S, Marugan J, Kim HY. Orphan GPR110 (ADGRF1) targeted by N-docosahexaenoylethanolamine in development of neurons and cognitive function. Nat Commun 7: 13123, 2016. doi: 10.1038/ncomms13123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Bassilana F, Nash M, Ludwig MG. Adhesion G protein-coupled receptors: opportunities for drug discovery. Nat Rev Drug Discov 18: 869–884, 2019. doi: 10.1038/s41573-019-0039-y. [DOI] [PubMed] [Google Scholar]
  • 142.Cazorla-Vázquez S, Engel FB. Adhesion GPCRs in kidney development and disease. Front Cell Dev Biol 6: 9, 2018. doi: 10.3389/fcell.2018.00009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Vizurraga A, Adhikari R, Yeung J, Yu M, Tall GG. Mechanisms of adhesion G protein-coupled receptor activation. J Biol Chem 295: 14065–14083, 2020. doi: 10.1074/jbc.REV120.007423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Demberg LM, Winkler J, Wilde C, Simon KU, Schon J, Rothemund S, Schoneberg T, Promel S, Liebscher I. Activation of adhesion G protein-coupled receptors: agonist specificity of stachel sequence-derived peptides. J Biol Chem 292: 4383–4394, 2017. doi: 10.1074/jbc.M116.763656. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Chen H, Nwe PK, Yang Y, Rosen CE, Bielecka AA, Kuchroo M, Cline GW, Kruse AC, Ring AM, Crawford JM, Palm NW. A forward chemical genetic screen reveals gut microbiota metabolites that modulate host physiology. Cell 177: 1217–1231.e18, 2019. doi: 10.1016/j.cell.2019.03.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Luo R, Jeong SJ, Jin Z, Strokes N, Li S, Piao X. G protein-coupled receptor 56 and collagen III, a receptor-ligand pair, regulates cortical development and lamination. Proc Natl Acad Sci USA 108: 12925–12930, 2011. doi: 10.1073/pnas.1104821108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Scholz N, Gehring J, Guan C, Ljaschenko D, Fischer R, Lakshmanan V, Kittel RJ, Langenhan T. The adhesion GPCR latrophilin/CIRL shapes mechanosensation. Cell Rep 11: 866–874, 2015. doi: 10.1016/j.celrep.2015.04.008. [DOI] [PubMed] [Google Scholar]
  • 148.Ward Y, Lake R, Yin JJ, Heger CD, Raffeld M, Goldsmith PK, Merino M, Kelly K. LPA receptor heterodimerizes with CD97 to amplify LPA-initiated RHO-dependent signaling and invasion in prostate cancer cells. Cancer Res 71: 7301–7311, 2011. doi: 10.1158/0008-5472.CAN-11-2381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Sharma AK, Mauer SM, Kim Y, Michael AF. Interstitial fibrosis in obstructive nephropathy. Kidney Int 44: 774–788, 1993. doi: 10.1038/ki.1993.312. [DOI] [PubMed] [Google Scholar]
  • 150.Promel S, Waller-Evans H, Dixon J, Zahn D, Colledge WH, Doran J, Carlton MB, Grosse J, Schoneberg T, Russ AP, Langenhan T. Characterization and functional study of a cluster of four highly conserved orphan adhesion-GPCR in mouse. Dev Dyn 241: 1591–1602, 2012. doi: 10.1002/dvdy.23841. [DOI] [PubMed] [Google Scholar]
  • 151.Veninga H, Becker S, Hoek RM, Wobus M, Wandel E, van der Kaa J, van der Valk M, de Vos AF, Haase H, Owens B, van der Poll T, van Lier RA, Verbeek JS, Aust G, Hamann J. Analysis of CD97 expression and manipulation: antibody treatment but not gene targeting curtails granulocyte migration. J Immunol 181: 6574–6583, 2008. doi: 10.4049/jimmunol.181.9.6574. [DOI] [PubMed] [Google Scholar]
  • 152.Fang W, Wang Z, Li Q, Wang X, Zhang Y, Sun Y, Tang W, Ma C, Sun J, Li N, Yi F. Gpr97 exacerbates AKI by mediating Sema3A signaling. J Am Soc Nephrol 29: 1475–1489, 2018. doi: 10.1681/ASN.2017080932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Ping YQ, Mao C, Xiao P, Zhao RJ, Jiang Y, Yang Z, An WT, Shen DD, Yang F, Zhang H, Qu C, Shen Q, Tian C, Li ZJ, Li S, Wang GY, Tao X, Wen X, Zhong YN, Yang J, Yi F, Yu X, Xu HE, Zhang Y, Sun JP. Structures of the glucocorticoid-bound adhesion receptor GPR97-Go complex. Nature 589: 620–626, 2021. doi: 10.1038/s41586-020-03083-w. [DOI] [PubMed] [Google Scholar]
  • 154.Schuster VL, Fejes-Toth G, Naray-Fejes-Toth A, Gluck S. Colocalization of H+-ATPase and band 3 anion exchanger in rabbit collecting duct intercalated cells. Am J Physiol Renal Physiol 260: F506–F517, 1991. doi: 10.1152/ajprenal.1991.260.4.F506. [DOI] [PubMed] [Google Scholar]
  • 155.DiBona GF, Kopp UC. Neural control of renal function. Physiol Rev 77: 75–197, 1997. doi: 10.1152/physrev.1997.77.1.75. [DOI] [PubMed] [Google Scholar]
  • 156.Bello-Reuss E. Effect of catecholamines on fluid reabsorption by the isolated proximal convoluted tubule. Am J Physiol Renal Physiol 238: F347–F352, 1980. doi: 10.1152/ajprenal.1980.238.5.F347. [DOI] [PubMed] [Google Scholar]
  • 157.Nord EP, Howard MJ, Hafezi A, Moradeshagi P, Vaystub S, Insel PA. Alpha 2 adrenergic agonists stimulate Na+-H+ antiport activity in the rabbit renal proximal tubule. J Clin Invest 80: 1755–1762, 1987. doi: 10.1172/JCI113268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Nakada J, Yamada H, Endou H. Evidence that alpha-1-adrenergic stimuli specifically increase gluconeogenesis of the isolated proximal convoluted tubule in the rat. Ren Physiol 9: 213–222, 1986. doi: 10.1159/000173086. [DOI] [PubMed] [Google Scholar]
  • 159.Chabardès D, Montégut M, Imbert-Teboul M, Morel F. Inhibition of α2-adrenergic agonists on AVP-induced cAMP accumulation in isolated collecting tubule of the rat kidney. Mol Cell Endocrinol 37: 263–275, 1984. doi: 10.1016/0303-7207(84)90096-0. [DOI] [PubMed] [Google Scholar]
  • 160.Krothapalli RK, Suki WN. Functional characterization of the alpha adrenergic receptor modulating the hydroosmotic effect of vasopressin on the rabbit cortical collecting tubule. J Clin Invest 73: 740–749, 1984. doi: 10.1172/JCI111267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Krothapalli RK, Duffy WB, Senekjian HO, Suki WN. Modulation of the hydro-osmotic effect of vasopressin on the rabbit cortical collecting tubule by adrenergic agents. J Clin Invest 72: 287–294, 1983. doi: 10.1172/JCI110968. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Iino Y, Troy JL, Brenner BM. Effects of catecholamines on electrolyte transport in cortical collecting tubule. J Membr Biol 61: 67–73, 1981. doi: 10.1007/BF02007632. [DOI] [PubMed] [Google Scholar]
  • 163.Terker AS, Yang CL, McCormick JA, Meermeier NP, Rogers SL, Grossmann S, Trompf K, Delpire E, Loffing J, Ellison DH. Sympathetic stimulation of thiazide-sensitive sodium chloride cotransport in the generation of salt-sensitive hypertension. Hypertension 64: 178–184, 2014. doi: 10.1161/HYPERTENSIONAHA.114.03335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Duan XP, Gu L, Xiao Y, Gao ZX, Wu P, Zhang YH, Meng XX, Wang JL, Zhang DD, Lin DH, Wang WH, Gu R. Norepinephrine-induced stimulation of Kir4.1/Kir5.1 is required for the activation of NaCl transporter in distal convoluted tubule. Hypertension 73: 112–120, 2019. doi: 10.1161/HYPERTENSIONAHA.118.11621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Itoh S, Carretero OA, Murray RD. Renin release from isolated afferent arterioles. Kidney Int 27: 762–767, 1985. doi: 10.1038/ki.1985.77. [DOI] [PubMed] [Google Scholar]
  • 166.Arif E, Nihalani D. Beta2-adrenergic receptor in kidney biology: a current prospective. Nephrology (Carlton) 24: 497–503, 2019. doi: 10.1111/nep.13584. [DOI] [PubMed] [Google Scholar]
  • 167.Hering L, Rahman M, Potthoff SA, Rump LC, Stegbauer J. Role of α2-adrenoceptors in hypertension: focus on renal sympathetic neurotransmitter release, inflammation, and sodium homeostasis. Front Physiol 11: 566871, 2020. doi: 10.3389/fphys.2020.566871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Noh MR, Jang HS, Kim J, Padanilam BJ. Renal sympathetic nerve-derived signaling in acute and chronic kidney diseases. Int J Mol Sci 21: 1647, 2020. doi: 10.3390/ijms21051647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Yung YC, Stoddard NC, Chun J. LPA receptor signaling: pharmacology, physiology, and pathophysiology. J Lipid Res 55: 1192–1214, 2014. doi: 10.1194/jlr.R046458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Lee JH, Kim D, Oh YS, Jun HS. Lysophosphatidic acid signaling in diabetic nephropathy. Int J Mol Sci 20: 2850, 2019. doi: 10.3390/ijms20112850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Li HY, Oh YS, Choi JW, Jung JY, Jun HS. Blocking lysophosphatidic acid receptor 1 signaling inhibits diabetic nephropathy in db/db mice. Kidney Int 91: 1362–1373, 2017. doi: 10.1016/j.kint.2016.11.010. [DOI] [PubMed] [Google Scholar]
  • 172.Colín-Santana CC, Avendaño-Vázquez SE, Alcántara-Hernández R, García-Sáinz JA. EGF and angiotensin II modulate lysophosphatidic acid LPA1 receptor function and phosphorylation state. Biochim Biophys Acta 1810: 1170–1177, 2011. doi: 10.1016/j.bbagen.2011.08.016. [DOI] [PubMed] [Google Scholar]
  • 173.Aisenberg WH, Huang J, Zhu W, Rajkumar P, Cruz R, Santhanam L, Natarajan N, Yong HM, De Santiago B, Oh JJ, Yoon AR, Panettieri RA, Homann O, Sullivan JK, Liggett SB, Pluznick JL, An SS. Defining an olfactory receptor function in airway smooth muscle cells. Sci Rep 6: 38231, 2016. doi: 10.1038/srep38231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Dalesio NM, Barreto OS, Pluznick JL, Berkowitz DE. Olfactory, taste, and photo sensory receptors in non-sensory organs: it just makes sense. Front Physiol 9: 1673, 2018. doi: 10.3389/fphys.2018.01673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Flegel C, Vogel F, Hofreuter A, Schreiner BS, Osthold S, Veitinger S, Becker C, Brockmeyer NH, Muschol M, Wennemuth G, Altmuller J, Hatt H, Gisselmann G. Characterization of the olfactory receptors expressed in human spermatozoa. Front Mol Biosci 2: 73, 2015. doi: 10.3389/fmolb.2015.00073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Lee SJ, Depoortere I, Hatt H. Therapeutic potential of ectopic olfactory and taste receptors. Nat Rev Drug Discov 18: 116–138, 2019. doi: 10.1038/s41573-018-0002-3. [DOI] [PubMed] [Google Scholar]
  • 177.Halperin Kuhns VL, Sanchez J, Sarver DC, Khalil Z, Rajkumar P, Marr KA, Pluznick JL. Characterizing novel olfactory receptors expressed in the murine renal cortex. Am J Physiol Renal Physiol 317: F172–F186, 2019. doi: 10.1152/ajprenal.00624.2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Pluznick JL, Protzko RJ, Gevorgyan H, Peterlin Z, Sipos A, Han J, Brunet I, Wan LX, Rey F, Wang T, Firestein SJ, Yanagisawa M, Gordon JI, Eichmann A, Peti-Peterdi J, Caplan MJ. Olfactory receptor responding to gut microbiota-derived signals plays a role in renin secretion and blood pressure regulation. Proc Natl Acad Sci USA 110: 4410–4415, 2013. doi: 10.1073/pnas.1215927110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Pluznick JL, Zou D-J, Zhang X, Yan Q, Rodriguez-Gil DJ, Eisner C, Wells E, Greer CA, Wang T, Firestein S, Schnermann J, Caplan MJ. Functional expression of the olfactory signaling system in the kidney. Proc Natl Acad Sci USA 106: 2059–2064, 2009. doi: 10.1073/pnas.0812859106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Shepard BD, Cheval L, Peterlin Z, Firestein S, Koepsell H, Doucet A, Pluznick JL. A renal olfactory receptor aids in kidney glucose handling. Sci Rep 6: 35215, 2016. doi: 10.1038/srep35215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Shepard BD, Koepsell H, Pluznick JL. Renal olfactory receptor 1393 contributes to the progression of type 2 diabetes in a diet-induced obesity model. Am J Physiol Renal Physiol 316: F372–F381, 2019. doi: 10.1152/ajprenal.00069.2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Armando I, Villar VA, Jose PA. Dopamine and renal function and blood pressure regulation. Compr Physiol 1: 1075–1117, 2011. doi: 10.1002/cphy.c100032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Cornil CA, Balthazart J, Motte P, Massotte L, Seutin V. Dopamine activates noradrenergic receptors in the preoptic area. J Neurosci 22: 9320–9330, 2002. doi: 10.1523/JNEUROSCI.22-21-09320.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Lazou A, Markou T, Zioga M, Vasara E, Efstathiou A, Gaitanaki C. Dopamine mimics the cardioprotective effect of ischemic preconditioning via activation of α1-adrenoceptors in the isolated rat heart. Physiol Res 55: 1–8, 2006. [DOI] [PubMed] [Google Scholar]
  • 185.Leedham JA, Pennefather JN. Selectivities of some agonists acting at α1- and α2-adrenoceptors in the rat vas deferens. J Auton Pharmacol 6: 39–46, 1986. doi: 10.1111/j.1474-8673.1986.tb00629.x. [DOI] [PubMed] [Google Scholar]
  • 186.Chen D, Stegbauer J, Sparks MA, Kohan D, Griffiths R, Herrera M, Gurley SB, Coffman TM. Impact of angiotensin type 1A receptors in principal cells of the collecting duct on blood pressure and hypertension. Hypertension 67: 1291–1297, 2016. doi: 10.1161/HYPERTENSIONAHA.115.06987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Kwon T-H, Nielsen J, Kim Y-H, Knepper MA, Frøkiær J, Nielsen S. Regulation of sodium transporters in the thick ascending limb of rat kidney: response to angiotensin II. Am J Physiol Renal Physiol 285: F152–F165, 2003. doi: 10.1152/ajprenal.00307.2002. [DOI] [PubMed] [Google Scholar]
  • 188.Nguyen MT, Lee DH, Delpire E, McDonough AA. Differential regulation of Na+ transporters along nephron during ANG II-dependent hypertension: distal stimulation counteracted by proximal inhibition. Am J Physiol Renal Physiol 305: F510–F519, 2013. doi: 10.1152/ajprenal.00183.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Ramkumar N, Kohan DE. Role of the collecting duct renin angiotensin system in regulation of blood pressure and renal function. Curr Hypertens Rep 18: 29, 2016. doi: 10.1007/s11906-016-0638-5. [DOI] [PubMed] [Google Scholar]
  • 190.Wang T, Giebisch G. Effects of angiotensin II on electrolyte transport in the early and late distal tubule in rat kidney . Am J Physiol Renal Physiol 271: F143–F149, 1996. doi: 10.1152/ajprenal.1996.271.1.F143. [DOI] [PubMed] [Google Scholar]
  • 191.Chung JJ, Goldstein L, Chen YJ, Lee J, Webster JD, Roose-Girma M, Paudyal SC, Modrusan Z, Dey A, Shaw AS. Single-cell transcriptome profiling of the kidney glomerulus identifies key cell types and reactions to injury. J Am Soc Nephrol 31: 2341–2354, 2020. doi: 10.1681/ASN.2020020220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Park S, Yang SH, Jeong CW, Moon KC, Kim DK, Joo KW, Kim YS, Lee JW, Lee H. RNA-Seq profiling of microdissected glomeruli identifies potential biomarkers for human IgA nephropathy. Am J Physiol Renal Physiol 319: F809–F821, 2020. doi: 10.1152/ajprenal.00037.2020. [DOI] [PubMed] [Google Scholar]
  • 193.Munk C, Mutt E, Isberg V, Nikolajsen LF, Bibbe JM, Flock T, Hanson MA, Stevens RC, Deupi X, Gloriam DE. An online resource for GPCR structure determination and analysis. Nat Methods 16: 151–162, 2019. doi: 10.1038/s41592-018-0302-x. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from American Journal of Physiology - Renal Physiology are provided here courtesy of American Physiological Society

RESOURCES