Loading

Annals of Clinical and Experimental Hypertension

Sorting Nexins: New Determinants for the Development of Hypertension

Review Article | Open Access

  • 1. Department of Cardiology, Daping Hospital, The Third Military Medical University, P.R.China
  • 2. Department of Nutrition, Daping Hospital, The Third Military Medical University, P.R.China
  • 3. Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, USA
  • 4. Department of Physiology, University of Maryland School of Medicine, USA
+ Show More - Show Less
Corresponding Authors
Van Anthony M. Villar, Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, 20 Penn St., Suite S003C, Baltimore, MD 21201, USA Jian Yang, Department of Cardiology, and Department of Nutrition Daping Hospital, The Third Military Medical University, Chongqing City, 400042, P.R. China
Abstract

G protein-coupled receptors (GPCRs) are serpentine seven-transmembrane receptors that mediate the cellular responses to a myriad of hormones and neurotransmitters. As such, many of these receptors are crucial to the regulation of important physiological processes, such as blood pressure and renal sodium transport. The trafficking and signal transduction of GPCRs, including the dopamine receptors, are tightly regulated to ensure the accuracy of the intracellular signal and to limit the specificity and extent of the cellular response. A growing body of evidence has shown that the sorting and intracellular trafficking of agonist-activated receptors, including the GPCRs, appear to be mediated by the sorting nexins, among a few other proteins. The sorting nexin family consists of a diverse group of cytoplasmic and membrane-associated proteins that contain the canonical phox homology (PX) domain and are involved in the various aspects of protein trafficking after receptor endocytosis. Perturbation of the process and/or deficiency of the proteins involved in GPCR trafficking and signaling may lead to receptor dysfunction, impaired homeostatic responses, and possibly disease state. In this review, we provide an overview of GPCR trafficking, highlight the sorting nexins that impact the GPCRs that are involved in blood pressure control, and expound on the mechanisms of how the loss of certain sorting nexins may eventually lead to hypertension.

Citation

Yang J, Armando I, Jones JE, Zeng C, Jose PA, et al. (2014) Sorting Nexins: New Determinants for the Development of Hypertension. Ann Clin Exp Hypertension 2(1): 1008.

Keywords

•    GPCRs
•    Sorting nexin
•    Blood pressure
•    Dopamine receptor
•    Receptor trafficking

ABBREVIATIONS

AT1R: Angiotensin II Type 1 Receptor; D1R, D2R, D3R, D4R, D5R: Dopamine D1-5 Receptor; EEA1: Early Endosome Antigen 1; EGFR: Epidermal Growth Factor Receptor; ENaC: Epithelial Na+ channel; GWAS: Genome-Wide Association Study; GPCR: G Protein-Coupled Receptor; GRK: GPCR Kinase; HEK293: Human Embryonic Kidney ells; hRPTCs: human Renal Proximal Tubule Cells; LAMP-1: Lysosomal-Associated Membrane Protein 1; (MMEP): Microcephaly Microphthalmia Ectrodactyly, Prognathism; NHE3: Na+ /H+ Exchanger isoform 3; PX: Phox homology; PDZ: Postsynaptic Density Protein-95/Discs-large, Zona-occludens-1; RA: Ras Association; SNX: Sorting Nexin; TGF: Transforming Growth Factor; TGN: Trans-Golgi Network; USP10: Ubiquitin-Specific Protease 10

INTRODUCTION

Hypertension is one of the most common and important health problems worldwide. Nearly 30% of middle-aged Americans have hypertension, but the prevalence is higher in non-Hispanic blacks and individuals >60 years of age (65%) [1]. In 2000, the worldwide prevalence of hypertension was estimated to be 26%, affecting approximately 1 billion people. It has been estimated that 29% of the world’s adult population, or 1.56 billion people, will have hypertension by the year of 2025 [2]. Genetic and environmental factors and their interaction determine an individual’s risk for hypertension [3,4]. Hypertension is a major risk factor for stroke, myocardial infarction, heart and kidney failure, and premature death globally [2,5,6].

G protein-coupled receptors (GPCRs) are a large and functionally diverse superfamily of cell-surface receptors that share a common architecture consisting of seven-transmembrane (TM) domains connected by extracellular and intracellular loops [7,8]. Upon ligand binding, GPCRs modulate a variety of cell functions by coupling to heterotrimeric G proteins and regulating downstream effectors such as adenylyl cyclases, phospholipases, protein kinases, and ion channels [9-12]. The signal transduction that follows ligand occupation of a GPCR is tightly regulated to limit the specificity and extent of the cellular response. GPCR-mediated signal transduction is rapidly dampened via receptor desensitization, or the waning of the responsiveness of the receptor to agonist stimulation with time [13,14]. GPCRs, including the dopamine and angiotensin II receptors, elicit cellular responses to a myriad of stimuli, play essential roles in human health, and have important clinical implications in various diseases [15,16]. By far, GPCRs have been intensively studied as a key factor in the basic physiology and pathophysiology of hypertension and its complications. Identifying GPCRs associated with blood pressure advances our understanding of blood pressure regulation and highlights potential and novel strategies for the prevention and treatment of hypertension.

GPCR TRAFFICKING AND SIGNAL TRANSDUCTION

Agonist activation of a GPCR results into two simultaneous processes, i.e., receptor signal transduction and receptor trafficking. GPCR agonism induces a conformational change of the receptor which is followed by the uncoupling of the receptor from its cognate trimeric G protein and its disassembly into Gα and Gβγ subunits. The Gα subunit either activates or inhibits the enzyme adenylyl cyclase (or other signaling enzymes) to either increase or decrease the production of cAMP (or other signal transducers). The Gβγ subunit recruits G protein-coupled receptor kinases (GRKs), which then selectively phosphorylate serine and threonine residues in the receptor to promote the binding of the β-arrestins. Once internalized, the GPCRs, in vesicles termed as early (sorting) endosomes, are sorted and follow divergent pathways (Figure 1). The receptors are: (a) sorted into recycling endosomes for their return to the cell membrane (resensitization, recycling, and re-insertion); (b) accumulated in late endosomes and are passed on to the lysosomes for their subsequent degradation; or (c) transported initially to the perinuclear endosomes (trans-Golgi network [TGN]) and then to the late endosomes for eventual lysosomal degradation [17-20]. Additional proteolytic mechanisms, such as the proteasomes and cell-associated endopeptidases, are also implicated in mediating the down-regulation of certain GPCRs [21,22].

The signal transduction that follows GPCR occupation by its ligand is highly regulated to ensure the specificity of the cellular response, both temporally and spatially. GPCR-mediated signal transduction can be attenuated rapidly through a process known as desensitization or through a slower process of downregulation after prolonged or repeated exposure to an agonist ligand. Rapid desensitization, or the waning of a receptor’s responsiveness to the agonist with time, is carried out via at least two complementary mechanisms, i.e., the functional uncoupling of receptors from G proteins and the sequestration and internalization of cell surface receptors [23]. These processes occur within a time frame of seconds (uncoupling) to minutes (internalization) to hours (down-regulation).

SORTING NEXINS

The sorting nexin (SNX) family consists of a diverse group of cytoplasmic and membrane-associated proteins that are involved in various aspects of receptor endocytosis and trafficking through the endosomes [24,25]. To date, SNXs have been identified across phyla, from yeast to mammals, and currently 10 yeast and 33 mammalian SNXs have been identified [26]. All SNX family members contain the canonical 100-130-amino acid phox homology (PX) domain, which is responsible for binding to specific phosphoinositides [20,26]. Some of the SNXs also possess a C-terminal Bin/Amphiphysin/Rvs (BAR) domain that is important for both dimerization with a similar SNX and detection of membrane curvature, an important feature for proteins that monitor changes in membrane architecture [27,28]. SNXs play pivotal roles in the whole pathway of endocytic trafficking, including endocytosis, endosomal sorting, and endosomal signaling [24-26].

Increasing number of studies have shown that SNXs are associated with diseases in which endosomal function is adversely perturbed, such as cancer, Alzheimer’s disease, Down’s syndrome, and hypertension [29-34]. Specifically, recent studies have shed light into the molecular mechanisms by which SNXs regulate GPCRs, such as the dopamine receptors [29,30], β1 - adrenergic receptor [35], and other receptors like the transferrin receptor and transforming growth factor beta (TGF-β)receptor I [36,37]. In this paper, we review the physiological actions of SNXs in the regulation of GPCRs that are involved in the regulation of blood pressure and discuss the possible mechanisms by which hypertension develops when the function of SNXs is perturbed.

RENAL DOPAMINE RECEPTORS

The kidney plays a major role in the long-term control of blood pressure and is the major organ involved in the regulation of sodium homoeostasis [38-43]. Many studies have focused on the abnormal renal handling of salt in the pathogenesis of hypertension [39,40,44-47]. The sodium retention in hypertension results from an enhanced sodium transport per se and/or a failure to respond appropriately to signals that decrease sodium transport. Humans with polygenic essential hypertension have increased sodium transport in the renal proximal tubule and medullary thick ascending limb, which are regulated by numerous hormones and humoral factors, including the dopamine and angiotensin II, which exert their effects via GPCRs [48-51].

Dopamine, a well-known neurotransmitter in the central nervous system, has also been characterized as an important modulator of blood pressure, sodium balance, and renal and adrenal function, and is relevant to the pathogenesis and/ or maintenance of hypertension [10,29,30,40,41,50,52-54]. Dopamine receptors are classified into the D1 - and D2 -like subtypes based on their structure and pharmacology. D1 -like receptors (D1 R and D5 R) couple to the stimulatory G protein GαS and stimulate adenylyl cyclase activity, whereas D2 -like receptors (D2 R, D3 R, and D4 R) couple to the inhibitory G protein Gαi /Gαo and inhibit adenylyl cyclase activity [41,50,55]. During conditions of moderate sodium balance, more than 50% of renal sodium excretion is regulated by the D1 -like receptors [41,50,56-59]. The D1 R increases cAMP production to a greater extent than the D5 R in renal proximal tubule cells [60]. However, the D5 R has a higher affinity for dopamine than the D1 R and exhibits constitutive activity [50,61]. Among the D2 -like dopamine receptors, the D3 R is the major receptor in the nephron and has 20 times higher affinity for dopamine than the D2 R.

All of the five dopamine receptor subtypes are expressed in the renal tubule and renal vasculature. Disruption of any of the dopamine receptor genes in mice results in hypertension, the pathogenesis of which is specific for each receptor subtype [10,50,61]. Disruption of the D1 R gene Drd1 in mice results in the development of hypertension [62], while that of the D2 R gene Drd2 leads to hypertension, in part, due to increased noradrenergic discharge [63]. The salt sensitivity of D1 R deficient mice remains to be determined but D2 R deficient mice may develop salt sensitivity [64]. Disruption of D3 R gene Drd3 induces a renin-dependent form of hypertension accompanied by failure to excrete a sodium load [65] that may be dependent on the genetic background [66], while Drd4 knockout mice exhibit hypertension, possibly through increased expression AT1 R [67] and also a failure to excrete a sodium load (unpublished data). Genetic ablation of the D5 R gene Drd5 in mice also results in hypertension, presumably caused by increased activity of the sympathetic nervous system due to activation of oxytocin, V1 vasopressin, and non-N-methyl D-aspartate receptors in the central nervous system [68,69], although increased renal angiotensin II type I receptor (AT1 R) protein expression [21,40,70] may also play a significant role. D5 R-deficient mice are salt-sensitive and have increased oxidative stress [21,71].

SORTING NEXINS AND GPCRS

SNX1 and D5R

There is now increasing evidence showing the importance of the D5 R in regulating blood pressure. The human D5 R gene DRD5 locus at 4p15.1–16.1 is linked to essential hypertension [72,73]. Disruption of Drd5 produces hypertension in (Drd5-/- ) mice and a high salt diet increases further the elevated blood pressure [21,40,71]. The renal expression of AT1 R and reactive oxygen species (ROS) are increased in Drd5-/-mice [21,40,71,74]. Heydorn et al first reported the association of the C-terminal tail of D5 R, but not of the other dopamine receptors, with SNX1 [75].

SNX1 was originally identified as a protein that interacts with the cytoplasmic sequences of the epidermal growth factor receptor (EGFR), including the tyrosine kinase domain and the adjacent lysosomal targeting signal [76,77]. SNX1 was the first mammalian sorting nexin to be characterized and is the ortholog of the yeast (vacuolar protein sorting) VPS5p, a protein involved in TGN trafficking. SNX1 can homodimerize or heterodimerize with SNX2 to form the membrane-targeting complex which, together with the cargo-recognition complex composed of Vps26, Vps29 and Vps35, forms the mammalian retromer, a protein complex that is involved in the retrograde trafficking between early endosomes and the TGN [78,79].

The human SNX1 consists of an N-terminal SNX region, a central PX domain, and a C-terminal BAR domain that binds to and/or induces membrane curvature via interactions with the lipid bilayer [80,81] (Figure 2A). It is distributed in both the plasma membrane and cytoplasm, where it exists in large complexes with other proteins [20]. Mutational analysis of SNX1 indicates that both an intact PX domain and an intact helical C-terminus are necessary for proper subcellular localization of SNX1 [82]. By way of “coincidence detection”, the tandem PX and BAR domains efficiently direct SNX1 to membrane microdomains characterized by the presence of phosphoinositides and high curvature [81] (Figure 2B).

The role for SNX1 in endosome-to-lysosome trafficking was first proposed based on studies in which SNX1 overexpression enhances EGFR degradation and SNX1 deletion or point mutations inhibits EGFR degradation [76,82]. The endogenous SNX1 PX domain can specifically bind to specific phosphoinositides that are highly enriched in early endosomal membranes, such as phosphatidylinositol-3-phosphate and phosphatidylinositol-3,5- bisphosphate [83]. SNX1 is implicated in endosome-to-lysosome sorting of cell surface receptors, including several tyrosine kinase receptors (EGFR, PDGFR, insulin receptor, transferrin receptor, and long form of the leptin receptor), and serine-threonine kinase receptors (TGF-βtype I and II receptors) [84,85]. A protein-protein interaction screen using SNX1 and a library of C-terminal tails from 59 GPCRs revealed that SNX1 is capable of interacting with at least 10 distinct GPCRs in vitro [86]. SNX1 is essential for sorting protease-activated receptor-1 (PAR1) to a distinct lysosomal degradative pathway that does not require retromer activity [87,88].

We have recently reported that in human renal proximal tubule cells (hRPTCs) and human embryonic kidney cells heterologously expressing the human D5 R (HEK293-hD5 R), SNX1 is essential for the trafficking and function of the D5 R [30]. SNX1, but not its homolog SNX2, colocalizes and coimmunoprecipitates with the D5 R in these cells and in renal proximal tubules in the human kidney. RNAi-mediated, kidneyspecific silencing of SNX1 results in the simultaneous impairment of three processes, i.e., (a) receptor internalization, (b) signal transduction, and (c) inhibition of AT1 R expression [30]. The failure of D5 R to internalize upon agonist stimulation prevents receptor resensitization, which is a prerequisite for sustained/ long-term receptor response. Moreover, SNX1 depletion prevents the D5 R signal transduction by inhibiting the binding of GTP to Gαs in exchange for GDP to activate the D5 R-coupled Gαs . This, in turn, leads to blunted cAMP response to agonist stimulation; Gαs cannot stimulate the enzyme adenylyl cyclase, which is needed to convert ATP to cAMP. Ultimately, SNX1 depletion leads to the failure of agonist-activated D5 R to inhibit sodium transport via Na+ ,K+ -ATPase and the up-regulation of AT1 R expression [30]; D5 R negatively regulates the expression of AT1 R [21,40,70]. Silencing of renal Snx1 results in increased AT1 R expression, elevated blood pressure, and impaired natriuretic response to D1 -like dopamine receptor agonist stimulation in two mouse strains, i.e., C57BL/6J and BALB/cJ mice[30]. We have proposed that SNX1 may initiate the sorting of the activated D5 R by tagging it for endocytosis and also serve as a scaffold or adaptor protein that facilitates the organization of the D5 R signaling complex [30].

SNX5 and D1R

SNX5 is a 404-amino acid residue protein that contains a central PX domain and large C-terminal domain predicted to include a BAR domain [89]. SNX5 is distributed in both the plasma membrane and the cytoplasm where it partially colocalizes with the early endosomal marker EEA1 [90,91], the late endosomal marker Rab7 [92], and with the lysosomal marker LAMP-1, suggesting a potential role in protein degradation. Compared with other SNXs, the SNX5 PX domain is unique in both its structure and ligand binding [93] since it can bind phosphatidylinositol3-phosphate and phosphatidylinositol-3,4-bisphosphate. This domain has been shown to be important in the ability of SNX5 to inhibit the degradation of EGFR [90].

SNX5 is expressed in many organs and cells. The highest levels of SNX5 mRNA are found in skeletal muscle and kidney, as well as in the MoLT-4 (T cell leukemia), SW80 (colon adenocarcinoma), and A549 (lung carcinoma) cell lines. Very little SNX5 mRNA is detected in the brain, placenta, lung, or liver, or in the HL60 (acute myelocytic leukemia), HeLa S3, and Raji (B cell leukemia) cell lines [94].

SNX5 was firstly identified by its interaction with the Fanconi anemia complementation group A protein [94]. Since then, SNX5 has been implicated in a myriad of cellular processes. SNX5 binds the clathrin heavy chain CHC22 and contributes to the specialization of CHC22 during myogenesis and muscle regeneration [95]. SNX5 colocalizes with the E3 ubiquitin ligase Mind bomb in early endosomal compartments and is important for its trafficking [96]. SNX5 also modulates the macropinocytic activity in primary mouse macrophages and influences the uptake and processing of soluble antigens [97,98]. SNX5 is necessary for the differentiation of alveolar epithelial type I cells. Disruption of SNX5 gene can lead to neonatal mortality caused by respiratory failure due to impaired differentiation of alveolar epithelial type I cells [99].

SNX5 can interact with other sorting nexins, especially with SNX6 and SNX1. SNX5 and the closely related SNX6 are the functional equivalents of Vps17p, a component of the yeast retromer complex [100], and make up a protein complex that mediates the endosome-to-TGN retrograde transport of the mannose 6-phosphate receptor [101]. Both SNX5 and SNX6 colocalize with SNX1 in early endosomes [102]. Although it cannot homodimerize, SNX5 can associate with SNX1 through its C-terminal region [90] to form SNX5/SNX1 heterodimers [92]. This interaction allows the trans-regulation of SNX5 and SNX1 by one another. SNX1 can attenuate the inhibitory effect of SNX5 on EGFR degradation [90], while suppression of SNX5 and/or SNX6 can result in a significant loss of SNX1, an effect that seems to result from post-translational regulation of the SNX1 [100]. However, other studies showed that SNX5 does not always interact with SNX1. SNX5 is selectively recruited to membrane ruffles of activated cells and is not associated with SNX1 [91]. The absence of SNX1 has no effect on SNX5 localization and macropinosome biogenesis in macrophages from SNX1 knockout mice [97]. A recent GWAS on a large cohort of European subjects revealed that the SNX5 single nucleotide polymorphism rs2328223 is associated increased LDL cholesterol levels [103].

We have recently identified the SNX5 as a novel binding partner for the C-terminus of D1 R [29]; its homolog SNX6 does not interact with D1 R. The D1 R is widely expressed in the kidney and plays a pivotal role in the regulation of sodium balance and maintenance of normal blood pressure [50,53]. Dopamine, via the D1 R, inhibits the activity of Na+ -K+ -ATPase in the basolateral membrane and the Na+ /H+ exchanger isoform 3 (NHE3) in the apical membrane of renal proximal tubule cells [50,51,52,54,104,105]. Disruption of the D1 R gene Drd1 in mice (Drd1-/-) results in increased blood pressure [10,50,61].

SNX5 and D1 R colocalize in renal epithelial cells and in the human kidney (mainly in proximal tubules) and the mouse kidney (proximal tubules) and brain (caudate nuclei and putamen). Agonist stimulation of the D1 R enhances the colocalization with SNX5 in renal tubule cells. siRNA-mediated depletion of endogenous SNX5 in hRPTCs impairs receptor internalization, markedly delays recycling, and blunts the increase in cAMP production in response to agonist stimulation. SNX5 may also restrain the GRK4 from accessing the phosphorylation sites of agonist-activated D1 R, which may explain an earlier observation that the initial 20 min of D1 R desensitization in hRPTCs is not caused by GRK4 [102]; GRK4 plays an important role in the homologous desensitization and proper orientation of D1 R in the plasma membrane. In spontaneously hypertensive rats (SHR), kidney-restricted subcapsular infusion of SNX5-specific siRNA further increases the systolic and diastolic blood pressure, which is associated with a decrease in sodium excretion [29]. Depletion of renal SNX5 in the normotensive BALB/c mice results in the development of hypertension (unpublished data).

Other sorting nexins

A few other members of the sorting nexin family have been implicated to be involved in certain processes that are germane to blood pressure regulation and water and electrolyte homeostasis.

SNX3: SNX3 is a predominantly cytosolic protein [85] that is highly expressed in peripheral leukocytes, spleen, heart, and skeletal muscle, and much less in the kidney. Disruption of SNX3 has been described in a patient with microcephaly, microphthalmia, ectrodactyly, prognathism (MMEP) phenotype [106], but not in others [107]. It facilitates the recycling of transferrin receptor and is required for the proper delivery of iron to erythroid progenitors; silencing of SNX3 results in anemia and hemoglobin defects in vertebrates due to impaired transferrin-mediated iron uptake and its accumulation in early endosomes [36].

The amiloride-sensitive epithelial Na+ channel (ENaC) in the distal nephron is involved in regulating Na+ levels in the extracellular fluid compartment. The hormone vasopressin regulates ENaC by promoting its translocation to the plasma membrane and regulating its expression levels. Boulkroun et al have reported that vasopressin increases the expression of ubiquitin-specific protease 10 (USP10), which deubiquitinylates and stabilizes SNX3 to increase the channel’s cell surface expression [108]. It has been suggested that USP10 and SNX3 act together to promote the export of ENaC via the secretory pathway to the plasma membrane, Alternatively, SNX3 may act as an adaptor for the channel and may engender its recycling via the retromer complex [108].

SNX19: SNX19 contains a central PX domain and non-BAR domains at the C-terminal region and its function is not clear. A few studies have shown that SNX19 is related to several diseases, including acute myeloid leukemia, thyroid tumors, and osteoarthritis [109,110,111]. However, SNX19 can interact with IA-2, a major autoantigen in type 1 diabetes and a regulator of insulin secretion [112]. Moreover, a variant of the SNX19 gene is associated with an increased risk of coronary heart disease [113].

SNX25: The full length SNX25 contains a typical PX domain, a PX-associated (PXA) domain, and a regulator of G protein Signaling (RGS) domain. The physiological role of SNX25 is unknown, although it is expressed in several tissues, including the brain and kidney [114]. SNX25 interacts with the TGF-β receptors and enhances the degradation of TGF-β receptor I via a clathrin-dependent endocytosis and endosome/lysosome degradation pathway [37]. The up-regulation of SNX25 is involved in the development of temporal lobe epilepsy [115]. SNX25 overexpression enhances the expression levels of both D1 R and D2 R, causes an increase in both D1 and D2 receptor-mediated signaling, and perturbs both endocytosis and recycling of the D2 R, but does not affect D1 R desensitization [116]. Depletion of endogenous SNX25 using siRNA causes a subsequent decrease in the D1 R and D2 R expression [114]. These observations suggest that SNX25 plays a role in the D1 R and D2 R trafficking through intracellular membrane compartments and regulates both receptor expression and signaling.

SNX27: SNX27 contains several domains, i.e., the PX domain, Ras association (RA) domain, and the postsynaptic density protein-95/Discs-large, Zona-occludens-1 (PDZ) domain, which functions as a scaffold to organize various proteins. It is primarily localized to the cytoplasm and partly to the plasma membrane. It is particularly enriched in vesicles of the recycling endocytic pathway, where it colocalizes with Rab11 and the transferrin receptors. Its vesicular localization is dependent on its PX domain. SNX27 is implicated in the molecular etiology of Down’s syndrome through its interaction with the ionotropic glutamate receptors (NMDA and AMPA receptors). Snx27-/- mice have severe neuronal deficits in the hippocampus and the cortex and exhibit defects in synaptic function, learning, and memory, and decreased NMDA and AMPA receptors [31], as well as growth retardation [117]. SNX27 also mediates the efficient recycling of the β1 -adrenergic receptor (β1 -AR) in monkey kidney-derived COS-1 cells [35] by linking to the retromer [118]. β1 -AR, the major subtype of adrenoceptor in cardiomyocytes, plays an important role in regulating cardiac output, an important determinant for blood pressure.

SUMMARY AND PERSPECTIVES

GPCRs are important for the regulation of blood pressure. The past few years have seen a rapidly growing appreciation of the importance of the sorting nexins in the biology and pathology of certain GPCRs, including the renal dopamine receptors. Recent studies have begun to highlight the pivotal roles of sorting nexins in the regulation of GPCR trafficking and signal transduction, and by extension, on water and electrolyte and blood pressure homeostasis. Further research using innovative silencing techniques and appropriate animal models will certainly lead to exciting advances in our understanding of the physiological functions of the sorting nexins and may demonstrate the sorting nexins as novel and crucial determinants for the pathogenesis of essential hypertension.

ACKNOWLEDGMENTS

These studies were supported, in part, by grants R01HL092196, R37HL023081, and R01DK090918 from the U.S. National Institutes of Health (NIH), and from the National Kidney Foundation of Maryland, and by grants 81100500, 30925018, 31130029, 81070559 from the National Natural Science Foundation of China.

REFERENCES

1. Egan BM, Zhao Y, Axon RN. US trends in prevalence, awareness, treatment, and control of hypertension, 1988-2008. JAMA. 2010; 303: 2043-2050.

2. Kearney PM, Whelton M, Reynolds K, Muntner P, Whelton PK, He J. Global burden of hypertension: analysis of worldwide data. Lancet. 2005; 365: 217-223.

3. Kato N, Takeuchi F, Tabara Y, Kelly TN, Go MJ, Sim X, et al. Meta-analysis of genome-wide association studies identifies common variants associated with blood pressure variation in east Asians. Nat Genet. 2011; 43: 531-538

4. Levy D, Ehret GB, Rice K, Verwoert GC, Launer LJ, Dehghan A, et al. Genome-wide association study of blood pressure and hypertension. Nat Genet. 2009; 41: 677-687.

5. S Burden of Disease Collaborators. The state of US health, 1990-2010: burden of diseases, injuries, and risk factors. JAMA. 2013; 310: 591- 608.

6. Lawes CM, Vander Hoorn S, Rodgers A; International Society of Hypertension. Global burden of blood-pressure-related disease, 2001. Lancet. 2008; 371: 1513-1518.

7. Alonso V, Friedman PA. Minireview: ubiquitination-regulated G protein-coupled receptor signaling and trafficking. Mol Endocrinol. 2013; 27: 558-572.

8. Jacoby E, Bouhelal R, Gerspacher M, Seuwen K. The 7 TM G-proteincoupled receptor target family. ChemMedChem. 2006; 1: 761-782.

9. Zamponi GW, Currie KP. Regulation of Ca(V)2 calcium channels by G protein coupled receptors. Biochim Biophys Acta. 2013; 1828: 1629- 1643.

10. Zeng C, Jose PA. Dopamine receptors: important antihypertensive counterbalance against hypertensive factors. Hypertension. 2011; 57: 11-17.

11. Premont RT, Gainetdinov RR. Physiological roles of G protein-coupled receptor kinases and arrestins. Annu Rev Physiol. 2007; 69: 511-534.

12. Armbruster BN, Roth BL. Mining the receptorome. J Biol Chem. 2005; 280: 5129-5132.

13. Schlegel W. Signal transduction via G protein coupled receptors: a personal outlook. J Recept Signal Transduct Res. 2010; 30: 493-499.

14. Conn PM, Ulloa-Aguirre A. Trafficking of G-protein-coupled receptors to the plasma membrane: insights for pharmacoperone drugs. Trends Endocrinol Metab. 2010; 21: 190-197.

15. Heng BC, Aubel D, Fussenegger M. An overview of the diverse roles of G-protein coupled receptors (GPCRs) in the pathophysiology of various human diseases. Biotechnol Adv. 2013; 31: 1676-1694.

16. Feigin ME. Harnessing the genome for characterization of G-protein coupled receptors in cancer pathogenesis. FEBS J. 2013; 280: 4729- 4738.

17. Burd CG. Physiology and pathology of endosome-to-Golgi retrograde sorting. Traffic. 2011; 12: 948-955.

18. Grant BD, Donaldson JG. Pathways and mechanisms of endocytic recycling. Nat Rev Mol Cell Biol. 2009; 10: 597-608.

19. Maxfield FR, McGraw TE. Endocytic recycling. Nat Rev Mol Cell Biol. 2004; 5: 121-132.

20. Worby CA, Dixon JE. Sorting out the cellular functions of sorting nexins. Nat Rev Mol Cell Biol. 2002; 3: 919-931.

21. Li H, Armando I, Yu P, Escano C, Mueller SC, Asico L, et al. Dopamine 5 receptor mediates Ang II type 1 receptor degradation via a ubiquitinproteasome pathway in mice and human cells. J Clin Invest. 2008; 118: 2180-2189.

22. Von Zastrow M. Mechanisms regulating membrane trafficking of G protein-coupled receptors in the endocytic pathway. Life Sci. 2003; 74: 217-224.

23. Ferguson SS. Evolving concepts in G protein-coupled receptor endocytosis: the role in receptor desensitization and signaling. Pharmacol Rev. 2001; 53: 1-24.

24. Cullen PJ, Korswagen HC. Sorting nexins provide diversity for retromer-dependent trafficking events. Nat Cell Biol. 2011; 14: 29-37.

25. Johannes L, Wunder C. The SNXy flavours of endosomal sorting. Nat Cell Biol. 2011; 13: 884-886.

26. Cullen PJ. Endosomal sorting and signalling: an emerging role for sorting nexins. Nat Rev Mol Cell Biol. 2008; 9: 574-582.

27. Masuda M, Mochizuki N. Structural characteristics of BAR domain superfamily to sculpt the membrane. Semin Cell Dev Biol. 2010; 21: 391-398.

28. Peter BJ, Kent HM, Mills IG, Vallis Y, Butler PJ, Evans PR, et al. BAR domains as sensors of membrane curvature: the amphiphysin BAR structure. Science. 2004; 303: 495-499.

29. Villar VA, Armando I, Sanada H, Frazer LC, Russo CM, Notario PM, et al. Novel role of sorting nexin 5 in renal D(1) dopamine receptor trafficking and function: implications for hypertension. FASEB J. 2013; 27: 1808-1819.

30. Villar VA, Jones JE, Armando I, Asico LD, Escano CS Jr, Lee H, et al. Sorting nexin 1 loss results in D5 dopamine receptor dysfunction in human renal proximal tubule cells and hypertension in mice. J Biol Chem. 2013; 288: 152-163.

31. Wang X, Zhao Y, Zhang X, Badie H, Zhou Y, Mu Y, et al. Loss of sorting nexin 27 contributes to excitatory synaptic dysfunction by modulating glutamate receptor recycling in Down’s syndrome. Nat Med. 2013; 19: 473-480.

32. Lee J, Retamal C, Cuitiño L, Caruano-Yzermans A, Shin JE, van Kerkhof P, et al. Adaptor protein sorting nexin 17 regulates amyloid precursor protein trafficking and processing in the early endosomes. J Biol Chem. 2008; 283: 11501-11508.

33. Nguyen LN, Holdren MS, Nguyen AP, Furuya MH, Bianchini M, Levy E, et al. Sorting nexin 1 down-regulation promotes colon tumorigenesis. Clin Cancer Res. 2006; 12: 6952-6959.

34. Williams R, Schlüter T, Roberts MS, Knauth P, Bohnensack R, Cutler DF. Sorting nexin 17 accelerates internalization yet retards degradation of P-selectin. Mol Biol Cell. 2004; 15: 3095-3105.

35. Nakagawa T, Asahi M. β1-adrenergic receptor recycles via a membranous organelle, recycling endosome, by binding with sorting nexin27. J Membr Biol. 2013; 246: 571-579.

36. Chen C, Garcia-Santos D, Ishikawa Y, Seguin A, Li L, Fegan KH, et al. Snx3 regulates recycling of the transferrin receptor and iron assimilation. Cell Metab. 2013; 17: 343-352.

37. Hao X, Wang Y, Ren F, Zhu S, Ren Y, Jia B, et al. SNX25 regulates TGF-β signaling by enhancing the receptor degradation. Cell Signal. 2011; 23: 935-946.

38. Herrera M, Coffman TM. The kidney and hypertension: novel insights from transgenic models. Curr Opin Nephrol Hypertens. 2012; 21: 171- 178.

39. Hall JE, Granger JP, do Carmo JM, da Silva AA, Dubinion J, George E, et al. Hypertension: physiology and pathophysiology. Compr Physiol. 2012; 2: 2393-2442.

40. Asico L, Zhang X, Jiang J, Cabrera D, Escano CS, Sibley DR, et al. Lack of renal dopamine D5 receptors promotes hypertension. J Am Soc Nephrol. 2011; 22: 82-89.

41. Banday AA, Lokhandwala MF. Dopamine receptors and hypertension. Curr Hypertens Rep. 2008; 10: 268-275.

42. Gomes P, Soares-da-Silva P. Dopamine acutely decreases type 3 Na(+)/H(+) exchanger activity in renal OK cells through the activation of protein kinases A and C signalling cascades. Eur J Pharmacol. 2004; 488: 51-59.

43. Moe OW. Sodium-hydrogen exchange in renal epithelia: mechanisms of acute regulation. Curr Opin Nephrol Hypertens. 1997; 6: 440-446.

44. Shimosawa T, Mu S, Shibata S, Fujita T. The kidney and hypertension: pathogenesis of salt-sensitive hypertension. Curr Hypertens Rep. 2012; 14: 468-472.

45. Rodriguez-Iturbe B, Franco M, Johnson RJ. Impaired pressure natriuresis is associated with interstitial inflammation in salt-sensitive hypertension. Curr Opin Nephrol Hypertens. 2013; 22: 37-44.

46. Strazzullo P, D’Elia L, Kandala NB, Cappuccio FP. Salt intake, stroke, and cardiovascular disease: meta-analysis of prospective studies. BMJ. 2009; 339: b4567.

47. Weinberger MH, Fineberg NS, Fineberg SE, Weinberger M. Salt sensitivity, pulse pressure, and death in normal and hypertensive humans. Hypertension. 2001; 37: 429-432.

48. Gonzalez-Vicente A, Garvin JL. Angiotensin II-induced hypertension increases plasma membrane Na pump activity by enhancing Na entry in rat thick ascending limbs. Am J Physiol Renal Physiol. 2013; 305: F1306-1314.

49. Aviv A, Hollenberg NK, Weder A. Urinary potassium excretion and sodium sensitivity in blacks. Hypertension. 2004; 43: 707-713.

50. Armando I, Villar VA, Jose PA. Dopamine and renal function and blood pressure regulation. Compr Physiol. 2011; 1: 1075-1117.

51. Doris PA. Renal proximal tubule sodium transport and genetic mechanisms of essential hypertension. J Hypertens. 2000; 18: 509- 519.

52. Hu MC, Di Sole F, Zhang J, McLeroy P, Moe OW. Chronic regulation of the renal Na(+)/H(+) exchanger NHE3 by dopamine: translational and posttranslational mechanisms. Am J Physiol Renal Physiol. 2013; 304: F1169-1180.

53. Harris RC, Zhang MZ. Dopamine, the kidney, and hypertension. Curr Hypertens Rep. 2012; 14: 138-143.

54. Aperia AC. Intrarenal dopamine: a key signal in the interactive regulation of sodium metabolism. Annu Rev Physiol. 2000; 62: 621- 647.

55. Cuevas S, Villar VA, Jose PA, Armando I. Renal dopamine receptors, oxidative stress, and hypertension. Int J Mol Sci. 2013; 14: 17553- 17572.

56. Hansell P, Fasching A. The effect of dopamine receptor blockade on natriuresis is dependent on the degree of hypervolemia. Kidney Int. 1991; 39: 253-258.

57. Chen CJ, Lokhandwala MF. An impairment of renal tubular DA-1 receptor function as the causative factor for diminished natriuresis to volume expansion in spontaneously hypertensive rats. Clin Exp Hypertens A. 1992; 14: 615-628.

58. Jose PA, Asico LD, Eisner GM, Pocchiari F, Semeraro C, Felder RA. Effects of costimulation of dopamine D1- and D2-like receptors on renal function. Am J Physiol. 1998; 275: R986-994.

59. Siragy HM, Felder RA, Howell NE, Chevalier RL, Peach MJ, Carey RM. Intrarenal dopamine acts at the dopamine-1 receptor to control renal function. J Hypertens Suppl. 1988; 6: S479-481.

60. Sanada H, Xu J, Watanabe H, Jose PA, Felder RA. Differential expression and regulation of dopamine-1(D1) and dopamine-5(D5) receptor function in human kidney. Am J Hypertens. 2000; 13: 156A.

61. Zeng C, Armando I, Luo Y, Eisner GM, Felder RA, Jose PA. Dysregulation of dopamine-dependent mechanisms as a determinant of hypertension: studies in dopamine receptor knockout mice. Am J Physiol Heart Circ Physiol. 2008; 294: H551-569.

62. Albrecht FE, Drago J, Felder RA, Printz MP, Eisner GM, Robillard JE, et al. Role of the D1A dopamine receptor in the pathogenesis of genetic hypertension. J Clin Invest. 1996; 97: 2283-2288.

63. Li XX, Bek M, Asico LD, Yang Z, Grandy DK, Goldstein DS, et al. Adrenergic and endothelin B receptor-dependent hypertension in dopamine receptor type-2 knockout mice. Hypertension. 2001; 38: 303-308.

64. Ueda A, Ozono R, Oshima T, Yano A, Kambe M, Teranishi Y, et al. Disruption of the type 2 dopamine receptor gene causes a sodiumdependent increase in blood pressure in mice. Am J Hypertens. 2003; 16: 853-858.

65. Asico LD, Ladines C, Fuchs S, Accili D, Carey RM, Semeraro C, et al. Disruption of the dopamine D3 receptor gene produces renindependent hypertension. J Clin Invest. 1998; 102: 493-498.

66. Luippold G, Piesch C, Osswald H, Mühlbauer B. Dopamine D3 receptor mRNA and renal response to D3 receptor activation in spontaneously hypertensive rats. Hypertens Res. 2003; 26: 855-861.

67. Bek MJ, Wang X, Asico LD, Jones JE, Zheng S, Li X, et al. AngiotensinII type 1 receptor-mediated hypertension in D4 dopamine receptordeficient mice. Hypertension. 2006; 47: 288-295.

68. Yang Z, Sibley DR, Jose PA. D5 dopamine receptor knockout mice and hypertension. J Recept Signal Transduct Res. 2004; 24: 149-164.

69. Hollon TR, Bek MJ, Lachowicz JE, Ariano MA, Mezey E, Ramachandran R, et al. Mice lacking D5 dopamine receptors have increased sympathetic tone and are hypertensive. J Neurosci. 2002; 22: 10801- 10810.

70. Zeng C, Yang Z, Wang Z, Jones J, Wang X, Altea J, et al. Interaction of angiotensin II type 1 and D5 dopamine receptors in renal proximal tubule cells. Hypertension. 2005; 45: 804-810.

71. Yang Z, Asico LD, Yu P, Wang Z, Jones JE, Escano CS, et al. D5 dopamine receptor regulation of reactive oxygen species production, NADPH oxidase, and blood pressure. Am J Physiol Regul Integr Comp Physiol. 2006; 290: R96-96R104.

72. Cai G, Cole SA, Freeland-Graves JH, MacCluer JW, Blangero J, Comuzzie AG. Principal component for metabolic syndrome risk maps to chromosome 4p in Mexican Americans: the San Antonio Family Heart Study. Hum Biol. 2004; 76: 651-665.

73. Allayee H, de Bruin TW, Michelle Dominguez K, Cheng LS, Ipp E, Cantor RM, et al. Genome scan for blood pressure in Dutch dyslipidemic families reveals linkage to a locus on chromosome 4p. Hypertension. 2001; 38: 773-778.

74. Lu Q, Yang Y, Villar VA, Asico L, Jones JE, Yu P, et al. D5 dopamine receptor decreases NADPH oxidase, reactive oxygen species and blood pressure via heme oxygenase-1. Hypertens Res. 2013; 36: 684-690.

75. Heydorn A, Søndergaard BP, Hadrup N, Holst B, Haft CR, Schwartz TW. Distinct in vitro interaction pattern of dopamine receptor subtypes with adaptor proteins involved in post-endocytotic receptor targeting. FEBS Lett. 2004; 556: 276-280.

76. Kurten RC, Cadena DL, Gill GN. Enhanced degradation of EGF receptors by a sorting nexin, SNX1. Science. 1996; 272: 1008-1010.

77. Kurten RC, Eddington AD, Chowdhury P, Smith RD, Davidson AD, Shank BB. Self-assembly and binding of a sorting nexin to sorting endosomes. J Cell Sci. 2001; 114: 1743-1756.

78. Hierro A, Rojas AL, Rojas R, Murthy N, Effantin G, Kajava AV, et al. Functional architecture of the retromer cargo-recognition complex. Nature. 2007; 449: 1063-1067.

79. Rojas R, Kametaka S, Haft CR, Bonifacino JS. Interchangeable but essential functions of SNX1 and SNX2 in the association of retromer with endosomes and the trafficking of mannose 6-phosphate receptors. Mol Cell Biol. 2007; 27: 1112-1124.

80. Zhong Q, Watson MJ, Lazar CS, Hounslow AM, Waltho JP, Gill GN. Determinants of the endosomal localization of sorting nexin 1. Mol Biol Cell. 2005; 16: 2049-2057.

81. Carlton J, Bujny M, Peter BJ, Oorschot VM, Rutherford A, Mellor H, et al. Sorting nexin-1 mediates tubular endosome-to-TGN transport through coincidence sensing of high- curvature membranes and 3-phosphoinositides. Curr Biol. 2004; 14: 1791-1800.

82. Zhong Q, Lazar CS, Tronchère H, Sato T, Meerloo T, Yeo M, et al. Endosomal localization and function of sorting nexin 1. Proc Natl Acad Sci U S A. 2002; 99: 6767-6772.

83. Cozier GE, Carlton J, McGregor AH, Gleeson PA, Teasdale RD, Mellor H, et al. The phox homology (PX) domain-dependent, 3-phosphoinositide-mediated association of sorting nexin-1 with an early sorting endosomal compartment is required for its ability to regulate epidermal growth factor receptor degradation. J Biol Chem. 2002; 277: 48730-48736.

84. Parks WT, Frank DB, Huff C, Renfrew Haft C, Martin J, Meng X, et al. Sorting nexin 6, a novel SNX, interacts with the transforming growth factor-beta family of receptor serine-threonine kinases. J Biol Chem. 2001; 276: 19332-19339.

85. Haft CR, de la Luz Sierra M, Barr VA, Haft DH, Taylor SI. Identification of a family of sorting nexin molecules and characterization of their association with receptors. Mol Cell Biol. 1998; 18: 7278-7287.

86. Heydorn A, Søndergaard BP, Ersbøll B, Holst B, Nielsen FC, Haft CR, et al. A library of 7TM receptor C-terminal tails. Interactions with the proposed post-endocytic sorting proteins ERM-binding phosphoprotein 50 (EBP50), N-ethylmaleimide-sensitive factor (NSF), sorting nexin 1 (SNX1), and G protein-coupled receptor-associated sorting protein (GASP). J Biol Chem. 2004; 279: 54291-54303.

87. Gullapalli A, Wolfe BL, Griffin CT, Magnuson T, Trejo J. An essential role for SNX1 in lysosomal sorting of protease-activated receptor-1: evidence for retromer-, Hrs-, and Tsg101-independent functions of sorting nexins. Mol Biol Cell. 2006; 17: 1228-1238.

88. Wang Y, Zhou Y, Szabo K, Haft CR, Trejo J. Down-regulation of protease-activated receptor-1 is regulated by sorting nexin 1. Mol Biol Cell. 2002; 13: 1965-1976.

89. Teasdale RD, Loci D, Houghton F, Karlsson L, Gleeson PA. A large family of endosome-localized proteins related to sorting nexin 1. Biochem J. 2001; 358: 7-16.

90. Liu H, Liu ZQ, Chen CX, Magill S, Jiang Y, Liu YJ. Inhibitory regulation of EGF receptor degradation by sorting nexin 5. Biochem Biophys Res Commun. 2006; 342: 537-546.

91. Merino-Trigo A, Kerr MC, Houghton F, Lindberg A, Mitchell C, Teasdale RD, et al. Sorting nexin 5 is localized to a subdomain of the early endosomes and is recruited to the plasma membrane following EGF stimulation. J Cell Sci. 2004; 117: 6413-6424.

92. Kerr MC, Lindsay MR, Luetterforst R, Hamilton N, Simpson F, Parton RG, et al. Visualisation of macropinosome maturation by the recruitment of sorting nexins. J Cell Sci. 2006; 119: 3967-3980.

93. Koharudin LM, Furey W, Liu H, Liu YJ, Gronenborn AM. The phox domain of sorting nexin 5 lacks phosphatidylinositol 3-phosphate (PtdIns(3)P) specificity and preferentially binds to phosphatidylinositol 4,5-bisphosphate (PtdIns(4,5)P2). J Biol Chem. 2009; 284: 23697-23707.

94. Otsuki T, Kajigaya S, Ozawa K, Liu JM. SNX5, a new member of the sorting nexin family, binds to the Fanconi anemia complementation group A protein. Biochem Biophys Res Commun. 1999; 265: 630-635

95. Towler MC, Gleeson PA, Hoshino S, Rahkila P, Manalo V, Ohkoshi N, et al. Clathrin isoform CHC22, a component of neuromuscular and myotendinous junctions, binds sorting nexin 5 and has increased expression during myogenesis and muscle regeneration. Mol Biol Cell. 2004; 15: 3181-3195.

96. Yoo KW, Kim EH, Jung SH, Rhee M, Koo BK, Yoon KJ, et al. Snx5, as a Mind bomb-binding protein, is expressed in hematopoietic and endothelial precursor cells in zebrafish. FEBS Lett. 2006; 580: 4409- 4416.

97. Lim JP, Teasdale RD, Gleeson PA. SNX5 is essential for efficient macropinocytosis and antigen processing in primary macrophages. Biol Open. 2012; 1: 904-914.

98. Lim JP, Wang JT, Kerr MC, Teasdale RD, Gleeson PA. A role for SNX5 in the regulation of macropinocytosis. BMC Cell Biol. 2008; 9: 58.

99. Im SK, Jeong H, Jeong HW, Kim KT, Hwang D, Ikegami M, et al. Disruption of sorting nexin 5 causes respiratory failure associated with undifferentiated alveolar epithelial type I cells in mice. PLoS One. 2013; 8: e58511.

100. Wassmer T, Attar N, Bujny MV, Oakley J, Traer CJ, Cullen PJ. A loss-offunction screen reveals SNX5 and SNX6 as potential components of the mammalian retromer. J Cell Sci. 2007; 120: 45-54.

101. Hara S, Kiyokawa E, Iemura S, Natsume T, Wassmer T, Cullen PJ, et al. The DHR1 domain of DOCK180 binds to SNX5 and regulates cation-independent mannose 6-phosphate receptor transport. Mol Biol Cell. 2008; 19: 3823-3835.

102. Watanabe H, Xu J, Bengra C, Jose PA, Felder RA. Desensitization of human renal D1 dopamine receptors by G protein-coupled receptor kinase 4. Kidney Int. 2002; 62: 790-798.

103. Global Lipids Genetics Consortium, Willer CJ, Schmidt EM, Sengupta S, Peloso GM, Gustafsson S, et al. Discovery and refinement of loci associated with lipid levels. Nat Genet. 2013; 45: 1274-1283.

104. Salyer S, Lesousky N, Weinman EJ, Clark BJ, Lederer ED, Khundmiri SJ. Dopamine regulation of Na+-K+-ATPase requires the PDZ-2 domain of sodium hydrogen regulatory factor-1 (NHERF-1) in opossum kidney cells. Am J Physiol Cell Physiol. 2011; 300: C425- 434.

105. Pedrosa R, Gomes P, Soares-da-Silva P. Distinct signalling cascades downstream to Gsalpha coupled dopamine D1-like NHE3 inhibition in rat and opossum renal epithelial cells. Cell Physiol Biochem. 2004; 14: 91-100.

106. Vervoort VS, Viljoen D, Smart R, Suthers G, DuPont BR, Abbott A, et al. Sorting nexin 3 (SNX3) is disrupted in a patient with a translocation t(6;13)(q21;q12) and microcephaly, microphthalmia, ectrodactyly, prognathism (MMEP) phenotype. J Med Genet. 2002; 39: 893-899.

107. Kumar RA, Everman DB, Morgan CT, Slavotinek A, Schwartz CE, Simpson EM. Absence of mutations in NR2E1 and SNX3 in five patients with MMEP (microcephaly, microphthalmia, ectrodactyly, and prognathism) and related phenotypes. BMC Med Genet. 2007; 8: 48.

108. Boulkroun S, Ruffieux-Daidié D, Vitagliano JJ, Poirot O, Charles RP, Lagnaz D, et al. Vasopressin-inducible ubiquitin-specific protease 10 increases ENaC cell surface expression by deubiquitylating and stabilizing sorting nexin 3. Am J Physiol Renal Physiol. 2008; 295: F889-900.

109. Kan A, Ikeda T, Saito T, Yano F, Fukai A, Hojo H, et al. Screening of chondrogenic factors with a real-time fluorescence-monitoring cell line ATDC5-C2ER: identification of sorting nexin 19 as a novel factor. Arthritis Rheum. 2009; 60: 3314-3323.

110. Tyybäkinoja A, Saarinen-Pihkala U, Elonen E, Knuutila S. Amplified, lost, and fused genes in 11q23-25 amplicon in acute myeloid leukemia, an array-CGH study. Genes Chromosomes Cancer. 2006; 45: 257-264.

111. Jacques C, Baris O, Prunier-Mirebeau D, Savagner F, Rodien P, Rohmer V, et al. Two-step differential expression analysis reveals a new set of genes involved in thyroid oncocytic tumors. J Clin Endocrinol Metab. 2005; 90: 2314-2320.

112. Hu YF, Zhang HL, Cai T, Harashima S, Notkins AL. The IA-2 interactome. Diabetologia. 2005; 48: 2576-2581.

113. Bare LA, Morrison AC, Rowland CM, Shiffman D, Luke MM, Iakoubova OA, et al. Five common gene variants identify elevated genetic risk for coronary heart disease. Genet Med. 2007; 9: 682-689.

114. Free RB, Namkung Y, Hazelwood LA, Sibley DR. Sorting nexin-25 interacts with D1 and D2 dopamine receptors to regulate receptor expression and signaling. FASEB J. 2010; 771: 8.

115. Du Y, Zou Y, Yu W, Shi R, Zhang M, Yang W, et al. Expression pattern of sorting Nexin 25 in temporal lobe epilepsy: a study on patients and pilocarpine-induced rats. Brain Res. 2013; 1509: 79-85.

116. Free RB, Hazelwood LA, Spalding HN, Cabrera DM, Sibley DR. Sorting nexin-25, a novel member of the dopamine receptor signalplex, upregulates D1 and D2 dopamine receptor expression in HEK293 cells. FASEB J. 2007; 21: 568.9.

117. Cai L, Loo LS, Atlashkin V, Hanson BJ, Hong W. Deficiency of sorting nexin 27 (SNX27) leads to growth retardation and elevated levels of N-methyl-D-aspartate receptor 2C (NR2C). Mol Cell Biol. 2011; 31: 1734-1747.

118. Temkin P, Lauffer B, Jäger S, Cimermancic P, Krogan NJ, von Zastrow M. SNX27 mediates retromer tubule entry and endosome-to-plasma membrane trafficking of signalling receptors. Nat Cell Biol. 2011; 13: 715-721.

Yang J, Armando I, Jones JE, Zeng C, Jose PA, et al. (2014) Sorting Nexins: New Determinants for the Development of Hypertension. Ann Clin Exp Hypertension 2(1): 1008.

Received : 12 Dec 2013
Accepted : 08 Jan 2014
Published : 09 Jan 2014
Journals
Annals of Otolaryngology and Rhinology
ISSN : 2379-948X
Launched : 2014
JSM Schizophrenia
Launched : 2016
Journal of Nausea
Launched : 2020
JSM Internal Medicine
Launched : 2016
JSM Hepatitis
Launched : 2016
JSM Oro Facial Surgeries
ISSN : 2578-3211
Launched : 2016
Journal of Human Nutrition and Food Science
ISSN : 2333-6706
Launched : 2013
JSM Regenerative Medicine and Bioengineering
ISSN : 2379-0490
Launched : 2013
JSM Spine
ISSN : 2578-3181
Launched : 2016
Archives of Palliative Care
ISSN : 2573-1165
Launched : 2016
JSM Nutritional Disorders
ISSN : 2578-3203
Launched : 2017
Annals of Neurodegenerative Disorders
ISSN : 2476-2032
Launched : 2016
Journal of Fever
ISSN : 2641-7782
Launched : 2017
JSM Bone Marrow Research
ISSN : 2578-3351
Launched : 2016
JSM Mathematics and Statistics
ISSN : 2578-3173
Launched : 2014
Journal of Autoimmunity and Research
ISSN : 2573-1173
Launched : 2014
JSM Arthritis
ISSN : 2475-9155
Launched : 2016
JSM Head and Neck Cancer-Cases and Reviews
ISSN : 2573-1610
Launched : 2016
JSM General Surgery Cases and Images
ISSN : 2573-1564
Launched : 2016
JSM Anatomy and Physiology
ISSN : 2573-1262
Launched : 2016
JSM Dental Surgery
ISSN : 2573-1548
Launched : 2016
Annals of Emergency Surgery
ISSN : 2573-1017
Launched : 2016
Annals of Mens Health and Wellness
ISSN : 2641-7707
Launched : 2017
Journal of Preventive Medicine and Health Care
ISSN : 2576-0084
Launched : 2018
Journal of Chronic Diseases and Management
ISSN : 2573-1300
Launched : 2016
Annals of Vaccines and Immunization
ISSN : 2378-9379
Launched : 2014
JSM Heart Surgery Cases and Images
ISSN : 2578-3157
Launched : 2016
Annals of Reproductive Medicine and Treatment
ISSN : 2573-1092
Launched : 2016
JSM Brain Science
ISSN : 2573-1289
Launched : 2016
JSM Biomarkers
ISSN : 2578-3815
Launched : 2014
JSM Biology
ISSN : 2475-9392
Launched : 2016
Archives of Stem Cell and Research
ISSN : 2578-3580
Launched : 2014
Annals of Clinical and Medical Microbiology
ISSN : 2578-3629
Launched : 2014
JSM Pediatric Surgery
ISSN : 2578-3149
Launched : 2017
Journal of Memory Disorder and Rehabilitation
ISSN : 2578-319X
Launched : 2016
JSM Tropical Medicine and Research
ISSN : 2578-3165
Launched : 2016
JSM Head and Face Medicine
ISSN : 2578-3793
Launched : 2016
JSM Cardiothoracic Surgery
ISSN : 2573-1297
Launched : 2016
JSM Bone and Joint Diseases
ISSN : 2578-3351
Launched : 2017
JSM Bioavailability and Bioequivalence
ISSN : 2641-7812
Launched : 2017
JSM Atherosclerosis
ISSN : 2573-1270
Launched : 2016
Journal of Genitourinary Disorders
ISSN : 2641-7790
Launched : 2017
Journal of Fractures and Sprains
ISSN : 2578-3831
Launched : 2016
Journal of Autism and Epilepsy
ISSN : 2641-7774
Launched : 2016
Annals of Marine Biology and Research
ISSN : 2573-105X
Launched : 2014
JSM Health Education & Primary Health Care
ISSN : 2578-3777
Launched : 2016
JSM Communication Disorders
ISSN : 2578-3807
Launched : 2016
Annals of Musculoskeletal Disorders
ISSN : 2578-3599
Launched : 2016
Annals of Virology and Research
ISSN : 2573-1122
Launched : 2014
JSM Renal Medicine
ISSN : 2573-1637
Launched : 2016
Journal of Muscle Health
ISSN : 2578-3823
Launched : 2016
JSM Genetics and Genomics
ISSN : 2334-1823
Launched : 2013
JSM Anxiety and Depression
ISSN : 2475-9139
Launched : 2016
Clinical Journal of Heart Diseases
ISSN : 2641-7766
Launched : 2016
Annals of Medicinal Chemistry and Research
ISSN : 2378-9336
Launched : 2014
JSM Pain and Management
ISSN : 2578-3378
Launched : 2016
JSM Women's Health
ISSN : 2578-3696
Launched : 2016
Clinical Research in HIV or AIDS
ISSN : 2374-0094
Launched : 2013
Journal of Endocrinology, Diabetes and Obesity
ISSN : 2333-6692
Launched : 2013
Journal of Substance Abuse and Alcoholism
ISSN : 2373-9363
Launched : 2013
JSM Neurosurgery and Spine
ISSN : 2373-9479
Launched : 2013
Journal of Liver and Clinical Research
ISSN : 2379-0830
Launched : 2014
Journal of Drug Design and Research
ISSN : 2379-089X
Launched : 2014
JSM Clinical Oncology and Research
ISSN : 2373-938X
Launched : 2013
JSM Bioinformatics, Genomics and Proteomics
ISSN : 2576-1102
Launched : 2014
JSM Chemistry
ISSN : 2334-1831
Launched : 2013
Journal of Trauma and Care
ISSN : 2573-1246
Launched : 2014
JSM Surgical Oncology and Research
ISSN : 2578-3688
Launched : 2016
Annals of Food Processing and Preservation
ISSN : 2573-1033
Launched : 2016
Journal of Radiology and Radiation Therapy
ISSN : 2333-7095
Launched : 2013
JSM Physical Medicine and Rehabilitation
ISSN : 2578-3572
Launched : 2016
Annals of Clinical Pathology
ISSN : 2373-9282
Launched : 2013
Annals of Cardiovascular Diseases
ISSN : 2641-7731
Launched : 2016
Journal of Behavior
ISSN : 2576-0076
Launched : 2016
Annals of Clinical and Experimental Metabolism
ISSN : 2572-2492
Launched : 2016
Clinical Research in Infectious Diseases
ISSN : 2379-0636
Launched : 2013
JSM Microbiology
ISSN : 2333-6455
Launched : 2013
Journal of Urology and Research
ISSN : 2379-951X
Launched : 2014
Journal of Family Medicine and Community Health
ISSN : 2379-0547
Launched : 2013
Annals of Pregnancy and Care
ISSN : 2578-336X
Launched : 2017
JSM Cell and Developmental Biology
ISSN : 2379-061X
Launched : 2013
Annals of Aquaculture and Research
ISSN : 2379-0881
Launched : 2014
Clinical Research in Pulmonology
ISSN : 2333-6625
Launched : 2013
Journal of Immunology and Clinical Research
ISSN : 2333-6714
Launched : 2013
Annals of Forensic Research and Analysis
ISSN : 2378-9476
Launched : 2014
JSM Biochemistry and Molecular Biology
ISSN : 2333-7109
Launched : 2013
Annals of Breast Cancer Research
ISSN : 2641-7685
Launched : 2016
Annals of Gerontology and Geriatric Research
ISSN : 2378-9409
Launched : 2014
Journal of Sleep Medicine and Disorders
ISSN : 2379-0822
Launched : 2014
JSM Burns and Trauma
ISSN : 2475-9406
Launched : 2016
Chemical Engineering and Process Techniques
ISSN : 2333-6633
Launched : 2013
Annals of Clinical Cytology and Pathology
ISSN : 2475-9430
Launched : 2014
JSM Allergy and Asthma
ISSN : 2573-1254
Launched : 2016
Journal of Neurological Disorders and Stroke
ISSN : 2334-2307
Launched : 2013
Annals of Sports Medicine and Research
ISSN : 2379-0571
Launched : 2014
JSM Sexual Medicine
ISSN : 2578-3718
Launched : 2016
Annals of Vascular Medicine and Research
ISSN : 2378-9344
Launched : 2014
JSM Biotechnology and Biomedical Engineering
ISSN : 2333-7117
Launched : 2013
Journal of Hematology and Transfusion
ISSN : 2333-6684
Launched : 2013
JSM Environmental Science and Ecology
ISSN : 2333-7141
Launched : 2013
Journal of Cardiology and Clinical Research
ISSN : 2333-6676
Launched : 2013
JSM Nanotechnology and Nanomedicine
ISSN : 2334-1815
Launched : 2013
Journal of Ear, Nose and Throat Disorders
ISSN : 2475-9473
Launched : 2016
JSM Ophthalmology
ISSN : 2333-6447
Launched : 2013
Journal of Pharmacology and Clinical Toxicology
ISSN : 2333-7079
Launched : 2013
Annals of Psychiatry and Mental Health
ISSN : 2374-0124
Launched : 2013
Medical Journal of Obstetrics and Gynecology
ISSN : 2333-6439
Launched : 2013
Annals of Pediatrics and Child Health
ISSN : 2373-9312
Launched : 2013
JSM Clinical Pharmaceutics
ISSN : 2379-9498
Launched : 2014
JSM Foot and Ankle
ISSN : 2475-9112
Launched : 2016
JSM Alzheimer's Disease and Related Dementia
ISSN : 2378-9565
Launched : 2014
Journal of Addiction Medicine and Therapy
ISSN : 2333-665X
Launched : 2013
Journal of Veterinary Medicine and Research
ISSN : 2378-931X
Launched : 2013
Annals of Public Health and Research
ISSN : 2378-9328
Launched : 2014
Annals of Orthopedics and Rheumatology
ISSN : 2373-9290
Launched : 2013
Journal of Clinical Nephrology and Research
ISSN : 2379-0652
Launched : 2014
Annals of Community Medicine and Practice
ISSN : 2475-9465
Launched : 2014
Annals of Biometrics and Biostatistics
ISSN : 2374-0116
Launched : 2013
JSM Clinical Case Reports
ISSN : 2373-9819
Launched : 2013
Journal of Cancer Biology and Research
ISSN : 2373-9436
Launched : 2013
Journal of Surgery and Transplantation Science
ISSN : 2379-0911
Launched : 2013
Journal of Dermatology and Clinical Research
ISSN : 2373-9371
Launched : 2013
JSM Gastroenterology and Hepatology
ISSN : 2373-9487
Launched : 2013
Annals of Nursing and Practice
ISSN : 2379-9501
Launched : 2014
JSM Dentistry
ISSN : 2333-7133
Launched : 2013
Author Information X