Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 Feb 1.
Published in final edited form as: FEBS J. 2021 Dec 26;290(3):600–619. doi: 10.1111/febs.16305

Sorting Nexins: Role in The Regulation of Blood Pressure

Juan Huang 1, Andrew C Tiu 2, Pedro A Jose 3, Jian Yang 1
PMCID: PMC9149145  NIHMSID: NIHMS1760583  PMID: 34847291

Abstract

Sorting nexins (SNXs) are a family of proteins that regulate cellular cargo sorting and trafficking, maintain intracellular protein homeostasis, and participate in intracellular signaling. SNXs are also important in the regulation of blood pressure, via several mechanisms. Aberrant expression and dysfunction of SNXs participate in the dysregulation of blood pressure. Genetic studies show correlation between SNX gene variants and response to antihypertensive drugs. In this review, we summarize the progress in SNX-mediated regulation of blood pressure, discuss the potential role of SNXs in the pathophysiology and treatment of hypertension, and propose novel strategies for the medical therapy of hypertension.

Keywords: sorting nexin, hypertension, trafficking, blood pressure, antihypertensive treatment

Introduction

Hypertension has become a serious public health problem [1]. It was predicted in 2005 that globally, in 2025, 29% of adult subjects (1.56 billion) may develop hypertension [2]. However, the incidence of hypertension has greatly exceeded this prediction. The global incidence of hypertension was 31.1%, or 1.39 billion people in 2010 [3]. The high prevalence of hypertension and its complications cause a huge economic burden worldwide [3,4].

The pathogenesis of essential hypertension is complex, involving inherited and external factors [59]. Many organs and systems including arteries, heart, kidneys, and nervous system play important roles in the development of hypertension. Numerous reports have demonstrated that lifestyle, including diet, exercise, sleep, and external factors, such as environmental pollution, affect the development, prevention, and treatment of hypertension [6,7]. Genetics and epigenetics are intricately associated with the pathogenesis and treatment of hypertension [8,9]. However, current studies have not totally uncovered the pathogenesis of hypertension, and thus, continuing investigation into the involvement of new factors and mechanisms are needed.

Sorting nexins (SNXs) are a family of proteins that participate in the trafficking of proteins in the cytoplasm and plasma membrane [10]. SNXs have been identified to modulate the entire endocytic process [1012]. Many reports have demonstrated that the deficiency of SNXs is associated with several diseases, including hypertension, in which protein sorting and trafficking are disturbed [1315]. In this review, we summarize the progress in SNX-mediated regulation of blood pressure, discuss the potential role of SNXs in the pathophysiology and treatment of hypertension, and propose novel strategies for the medical therapy of hypertension.

The subfamilies of SNXs

Ten subtypes in yeast and 33 subtypes in mammals have been identified as members of the SNX family [16]. The SNX members are categorized into five subfamilies based on their structural domains [12,17]: SNX-PX subfamily, SNX-BAR (Bin/Amphiphysin/Rvs) subfamily, SNX-FERM (protein 4.1/ezrin/radixin/moesin) subfamily, SNX-PXA (PX-associated domain A)-RGS (regulator of G-protein signaling domain)-PXC (PX-associated domain C) subfamily, and others. Basically, all SNXs share a phagocyte oxidase (phox) homology (PX) domain, containing the evolutionarily canonical 100–130 amino acids [17]. (1) The SNX-PX subfamily has ten members (SNX3/10/11/12/16/20/21/22/24/29). Their common PX domain interacts with phosphatidylinositols (PIs), such as phosphatidylinositol-3-phosphate (PI3P), which is well expressed in organelle membranes [18,19]. (2) The SNX-BAR subfamily has twelve members (SNX1/2/4/5/6/7/8/9/18/30/32/33). They possess a special BAR domain in the C-terminal and common PX domains [20]. (3) The SNX-FERM subfamily has three members (SNX17/27/31). They have a FERM domain, with an atypical tertiary structure in the C-terminal and PX domains [21,22]. Specifically, SNX27 has an N-terminal postsynaptic density 95/discs large/zonula occludens (PDZ) domain [23]. (4) The SNX-PXA-RGS-PXC subfamily has four members (SNX13/14/19/25). They have a PXA-RGS-PX-PXC or PXA-PX-PXC domain in the C-terminal domain [24,25]. The other unique SNX subfamily includes SNX15, SNX23, SNX26, and SNX28 [17,26,27].

Biological functions of SNXs

The most basic role of SNXs is being a vital constituent of the retromer [2830]. The retromer is a heteropentameric complex composed of two associated but distinct subcomplexes: a membrane-associated SNX dimeric subcomplex and a heterotrimer that includes VPS26, VPS29, and VPS35 [28]. The retromer core complex is an endosomal scaffold, that mediates the recycling or trafficking of intracellular proteins, such as G protein-coupled receptors (GPCRs) and other cargos from endosomes to either the trans-Golgi network or the plasma membrane [29,30]. The retromer interacts with three SNX subfamily members, including SNX3 from the SNX-PX subfamily, SNX-BAR subfamily members, and SNX27 from the SNX-FERM subfamily. The retromer VPS35 and a heterodimer, such as SNX1/5 and SNX2/6, participate in the insulin-like growth factor 2 receptor retrieval [31]. The SNX27-retromer complex participates in the regulation of transmembrane cargos, including the parathyroid hormone receptor; the SNX27 and parathyroid receptor interaction occurs with their the PDZ domains. However, the overall rate of recycling of the parathyroid hormone does not involve the PDZ domain but rather through a direct interaction of the retromer [32]. The SNX3-retromer regulates the membrane distribution and function of proteins associated with polycystic kidney disease [33]. However, SNX3 also mediates retromer-independent functions, such as facilitating tubular endosomal recycling of intracellular cargoes [34]. These reports indicate that the retromer complex and associated SNXs comprise a vital cellular trafficking machinery, which plays an important role in endosomal protein sorting.

SNXs are important in the regulation of the routing of cargoes, such as mitochondria, endoplasmic reticulum, peroxisomes, and cytoplasmic protein aggregates [35], via endocytosis, autophagy, and other trafficking pathways [36]. Endocytosis mediates the cellular uptake of extracellular components through membrane invagination [37]. SNX9 promotes podocin endocytosis associated with the chemotherapeutic drug adriamycin-induced podocyte injury and participates in clathrin-dependent and -independent endocytosis and cargo trafficking [38,39]. SNX33 modulates the endocytosis and alpha-secretase cleavage of amyloid precursor proteins [40]. SNXs also modulate the activity and/or function of cargoes by regulating varying trafficking pathways, but not their expression. For example, SNX1 is important for the trafficking of group 1 metabotropic glutamate receptors by directing them into a slow recycling pathway which allows receptor resensitization. The absence of SNX1 results in faster recycling of group 1 metabotropic receptors, preventing their resensitization [41]. In intestinal epithelial cells, SNX27 regulates the targeting of sodium-hydrogen exchanger type 3 (NHE3) at the plasma membrane by promoting the exocytosis of NHE3 from early endosome to the plasma membrane; SNX27 silencing decreases the plasma membrane activity and expression of NHE3 activity but not its total expression [42].

SNXs are also important in the regulation of the expression of intracellular proteins, via the proteasomal and lysosomal pathways [35]. In colorectal cancer cells, SNX16 activates c-Myc signaling by suppressing ubiquitin-mediated proteasomal degradation of the eukaryotic translation elongation factor 1A2 [43]. SNX1 modulates the protein decay of epidermal growth factor receptor (EGFR) by sorting it to lysosomes [44,45]. SNX11 facilitates the transport of transient receptor potential cation channel subfamily V member 3 into lysosomes for its decay [46]. SNX5 regulates the degradation of vesicular acetylcholine transporter, via the lysosomal pathway [47]. However, there are inconsistent reports on the effects of SNXs in the regulation of protein decay. For example, SNXs are also involved in the prevention of protein degradation. SNX10 restrains the development of colorectal cancer by inhibiting the trafficking of SRC, a non-receptor tyrosine kinase, to lysosomes where it would be degraded [48]. SNX17 by interacting with β1-integrin, causes its trafficking to the membrane, preventing its lysosomal degradation [49]. SNX27 interacts with the retromer VPS26, leading to the retardation of the transport of cargos to lysosomes [50]. These suggest that SNXs not only promote intracellular protein degradation but also are involved in the prevention of protein degradation.

SNXs can directly determine the destination of cells by regulating vital signaling molecules or pathways. For example, SNX9 inhibits the proliferation of immortalized autosomal dominant polycystic kidney disease cells by activating the Hippo-yes-associated protein signaling pathway [51]. SNX7 exerts its anti-apoptotic effect via Fas-associated death domain-like IL-1 converting enzyme-like inhibitory protein/caspase 8 pathway during the embryonic liver development [52]. SNX5 is essential for virus-induced autophagy by interacting with beclin 1 and autophagy-related gene (ATG) 14-containing class III phosphatidylinositol-3-kinase complex 1 [53]. SNX4 mediates the recycling of ATG9A, a membrane spanning ATG protein, from endolysosomes and autolysosomes to early endosomes, which is essential for autophagy [54]. SNX5 facilitates ferroptosis, a programmed cell death that is dependent on iron, in Parkinson’s disease models. Conversely, SNX5 depletion reduces dopaminergic neuronal ferroptosis in 6-OHDA-lesioned PC12 cells, a rat pheochromocytoma cell line [55]. Thus, current studies demonstrate that SNXs are involved in multiple cellular biological activities, including cell proliferation, apoptosis, autophagy, and ferroptosis.

SNXs have other physiological functions. SNXs directly participate in the intracellular signaling cascade. SNX10 controls mTOR activation, via the modulation of amino acid metabolism [56]. SNX9 makes a distinction between transforming growth factor β (TGF-β)-activated Smad3 from Smad2 and Smad4 to promote rapid nuclear delivery [57,58]. Our studies also found that SNX5 siRNA treatment reduces the amount of insulin receptors in human renal proximal tubule cells (hRPTCs), decreasing the levels of insulin receptor substrate-1 (IRS-1) and phospho-protein kinase B, two vital molecules in the insulin receptor-mediated signaling cascade [59]. SNXs are also involved in other intracellular functions, including restricting endolysosome motility, regulating the saturated fatty acid metabolism and endoplasmic reticulum homeostasis, and modulating cellular lipid deposition, among others [6062].

Regulation of blood pressure by SNXs

SNXs regulate blood pressure by three mechanisms: renal sodium excretion, vasoconstriction and vascular remodeling, and insulin sensitivity. SNXs and their intracellular functions associated with the regulation of blood pressure are presented in Table 1.

Table 1.

Summary of SNXs and their intracellular functions associated with the regulation of blood pressure

Isoform Cell types Cargos Intracellular distribution Intracellular physiological functions References
SNX1 hRPTCs D5R Distributed to a greater extent in the cytoplasm than at the cell membrane in hRPTCs Required for D5R endocytosis upon agonist stimulation;
Snx1 silencing (siRNA) in hRPTCs impairs D5R internalization, GTP binding, and function (stimulation of cAMP production and inhibition of sodium transport)
[89]
VSMCs AT1R Distributed in the cytoplasm and at the cell membrane Snx1 knockdown increases AT1R protein expression and its mediation of Ca2+ signaling [97]
SNX3 HEK-293 cells, mCCD cells ENaC Increases ENaC cell surface expression [124]
SNX4 COS-7 cells ENaC Distributed in early endosomes and recycling endosomes Promotes the endocytosis of ENaC [121]
SNX5 HEK-293 cells D1R Expressed at the plasma membrane and in the cytoplasm Snx5 silencing (siRNA) in hRPTCs increases agonist-activated D1R phosphorylation, prevents D1R internalization and cAMP response, and delays D1R receptor recycling to the plasma membrane [77]
hRPTCs IR Snx5 silencing (siRNA) in hRPTCs decreases IR expression and abundance of p-IRS and p-PKB [59]
hRPTCs IDE Snx5 silencing (siRNA) in hRPTCs decreases IDE expression and activity [134]
SNX9 HUVECs β1-integrins Required for tube formation in HUVECs and regulates recycling of β1-integrins from endosomes to the plasma membrane [103]
3T3L1 adipocytes IR GLUT4 Expressed to a greater extent in the cytosol than at the plasma membrane Regulates the IR trafficking and GLUT4 translocation to the plasma membrane induced by insulin [139]
SNX10 Mouse BMDMs, human PBMCs CD36 SNX10 deficiency results in the polarization of macrophages towards the anti-inflammatory M2 phenotype, and inhibition of foam cell formation by interrupting the internalization of CD36 [110,111]
SNX11 HEK-293T cells, HeLa cells TRPV3 Distributed throughout the cytoplasm Promotes TRPV3 trafficking from the plasma membrane to lysosomes for degradation [46]
SNX17 mouse fibroblasts β1-integrins Distributed in early endosomes and recycling endosomes Required for β1-integrin recycling and stabilization, and regulation of integrin-mediated cell function [49]
HUVECs P-selectin Expressed in the cytosol Promotes the endocytosis of P-selectin, and retards its movement into lysosomes [112]
SNX19 hRPTCs D1R Distributed in the cytoplasm and at the plasma cell membrane Silencing Snx19 (siRNA) in hRPTCs impairs D1R-mediated increase in cAMP production and inhibition of basolateral sodium transport [80]
pancreatic β cells DCVs Silencing Snx19 (siRNA) in mouse pancreatic cells decreases insulin secretion and the number of DCVs;
SNX19 interacts with IA-2 to initiate the pre-apoptotic state of pancreatic β cells
[127129]
SNX25 HEK-293T cells D1R, D2R Localized in distinct clusters near the plasma membrane Overexpression of SNX25 increases the expression of D1R and D2R, enhances receptor-mediated signaling, and perturbs both endocytosis and recycling of D2R;
Silencing Snx25 (siRNA) decreases D1R and D2R expression
[83,84]
SNX27 Caco2 cells DRA Localized in early endosomes Mediates DRA recycling to facilitate its activity in lipid rafts at the cell plasma membrane [123]
SK-CO15 cells NHE3 Mainly localized intracellularly Promotes cell surface expression of NHE3 [42]
Rat IMCD cells AQP2 Mainly localized intracellularly Regulates the autophagy-lysosomal degradation of AQP2 protein [125]
mouse and human β cells GLP-1R Distributed to a greater extent in the cytoplasm than at the plasma membrane Regulates GLP-1R trafficking and signaling [130]
HeLa cells GLUT1 Modulates GLUT1 lysosomal degradation and plasma membrane levels;
Snx27 silencing (shRNA) decreases GLUT1 recycling to the cell plasma membrane
[50]
human SGBS cells GLUT4 Distributed to a greater extent in the cytoplasm than at the cell membrane, but insulin recruits SNX27 to the plasma membrane Snx27 silencing (shRNA) decreases GLUT4 expression [141]
SNX31 mouse fibroblasts β1-integrins Distributed in early endosomes and recycling endosomes Regulates β1-integrins surface levels and stability by binding to its tails [105]

Abbreviations: AQP2, aquaporin-2 protein; AT1R, ang II type 1 receptor; BMDMs, bone marrow derived macrophage; D1R, dopamine D1 receptor; D2R, dopamine D2 receptor; D5R, dopamine D5 receptor; DCVs, dense core vesicles; DRA, down-regulation adenoma protein (aka SLC26A3 and chloride anion exchanger); ENaC, epithelial Na+ channel; GLP-1R, glucagon-like peptide type 1 receptor; GLUT1, glucose transporter 1; GLUT4, glucose transporter type 4; hRPTCs, human renal proximal tubule cells; HUVECs, human umbilical vein endothelial cells; IA-2, islet antigen-2; IDE, insulin-degrading enzyme; IMCD, inner medullary collecting duct; IR, insulin receptor; mCCD, mouse cortical collecting duct; PBMCs, peripheral blood mononuclear cells; p-IRS, phosphorylated insulin receptor substrate 1; p-PKB, phosphorylated protein kinase B; SGBS, Simpson-Golabi-Behmel syndrome; SNX, sorting nexin; TRPV3, transient receptor potential cation channel subfamily V member, Usp10, ubiquitin-specific protease 10; VSMCs, vascular smooth muscle cells.

Renal SNXs and blood pressure

The kidney is the main organ involved in the regulation of sodium balance and blood pressure [63]. Essential hypertension may be initiated by an increase in renal sodium reabsorption and an inappropriate decrease in renal sodium reabsorption and increase in sodium excretion with the increase in blood pressure. This impaired pressure natriuresis in hypertension is, in part, caused by a predominance of the autocrine, paracrine, and endocrine factors that increase renal sodium transport, such as angiotensin II, over the autocrine, paracrine, and endocrine factors that decrease renal sodium transport, such as dopamine [6366].

Dopamine exerts its natriuretic effects through the occupation of renal several receptor subtypes, which are categorized into D1-like (D1R/D5R) and D2-like receptor (D2R/D3R/D4R) subtypes [6769]. In hypertensive states, the expressions of renal dopamine receptors are reduced, which cause impaired sodium excretion, and thus resulting in a positive sodium balance [7072]. SNXs and dopamine receptors in the kidney interact to regulate blood pressure.

SNX-mediated regulation of renal D1R

There are abundant D1Rs in the kidney, which participate in the regulation of sodium excretion [73]. During normal or moderately increased NaCl intake, urinary sodium excretion is increased due, in part, to increased dopamine generation and enhanced D1-like receptor activation in the kidney [74]. The knockout of the Drd1 gene in mice promotes the development of hypertension [75,76].

The D1R is regulated by SNX5. Our previous studies showed that SNX5 and D1R colocalize in several segments of the renal tubule, including the proximal and distal tubules, of humans and rats [77]. We also found their localization in the brain, showing that the interaction between SNX5 and D1R is not restricted to the kidney [77]. Treatment of hRPTCs with fenoldopam, an agonist for both D1R and D5R, promotes the endocytosis of D1R and changes its intracellular distribution, as well as SNX5 [77]. Further studies showed that SNX5 not only regulates the endocytosis and cyclic adenosine monophosphate (cAMP) response induced by stimulation of D1R, but also the transport of D1R back to cell membrane, involving its C-terminus [78,79]. SNX5 also regulates renal D1R function in hypertensive animals. Renal-selective depletion of Snx5 aggravates the already elevated blood pressure in spontaneously hypertensive rats (SHRs), which is not observed in vehicle-infused SHRs. The further increase in blood pressure in SHRs with Snx5 knockdown is accompanied by impaired natriuresis [77].

The D1R in the kidney is also regulated by other SNXs. In hRPTCs, SNX19 colocalizes with D1R in the cytoplasm and at the plasma membrane; activation of D1Rs with fenoldopam causes their endocytosis [80]. In the basal state, in the mouse kidney, as in hRPTCs, the colocalization between SNX19 and D1R is also observed at the plasma membrane of renal proximal tubules (RPTs); the intravenous infusion of fenoldopam increased the colocalization of SNX19 and D1R in the cytosol of RPTs. The SNX19 interaction with D1R is specific because the D5R does co-immunoprecipitate with SNX19 in RPTs [80]. SNX19 participates in the palmitoylation of D1R, needed for its targeting to lipid rafts, which are specialized plasma membrane microdomains enriched by sterols and sphingolipids [8082]. The SNX19-mediated regulation of cellular D1R localization is important in the function of renal D1R in cell and animal experiments. Snx19 depletion in hRPTCs decreases the fenoldopam-mediated cAMP generation and inhibition of sodium pump activity. Snx19 depletion in normal salt diet-fed mice decreases renal D1R expression, leading to an increase in systolic blood pressure. Renal-selective overexpression of Drd1 normalizes the systolic blood pressure in these mice with kidneys depleted of Snx19, which is not observed in mice overexpressed with mutant D1R [80]. SNX25 also regulates D1R in the kidney. Overexpression of Snx25 in HEK 293 cells heterologously expressing D1R and D2R increases the expression of D1R and D2R, activates the downstream molecules, and disturbs the endocytosis and recycling of D2R. Conversely, the expressions of D1R and D2R are reduced in cells lacking Snx25 [83,84]. Thus, current studies show that renal D1R is regulated by some SNXs, specifically SNX5, SNX19, and SNX25. In addition, Rao et al reported that SNX13 may interact with D1R in the modulation of renal albumin excretion [85], which may be predictive of the risk of cardiovascular events in patients with essential hypertension [86].

SNX1-mediated regulation of renal D5R.

D5R, the other dopamine D1-like receptor subtype in humans, is also involved in the regulation of blood pressure [87]. The D5R is constitutively active and has a higher affinity for dopamine than the DlR [88]. Our previous studies showed that SNX1 regulates D5R trafficking and its functions in human and mouse kidney cells [89,90]. SNX1, as well as the retromer, including SNX1/2, participates in intracellular cargo transport [91,92].

D1R and D5R can be clearly distinguished from each other by SNX1, which binds strongly to the D5R but not D1R C-terminal tail [93]. In the human kidney, SNX1 is well expressed in the RPT and glomerulus. In hRPTCs, SNX1 colocalizes with D5R that is mostly evident at the cell membrane [89]. Treatment of hRPTCs with fenoldopam causes D5R endocytosis, and subsequently accelerates its translocation to the membrane. Snx1 depletion also impairs D5R-mediated physiological functions, including inhibition of renal sodium pump activity and impairment of cAMP generation [89]. These interactions in vitro are reflected in in vivo studies. Snx1 knockout (Snx1−/−) or renal Snx1 knockdown, using Snx1-specific siRNA, in mice increases systolic blood pressure and blunts the natriuretic response to fenoldopam [89,90]. The impaired natriuretic response to dopamine when Snx1 is silenced is related to the increased renal expression of sodium exchangers, pump, and cotransporters such as NHE3, Na+-K+-ATPase, and the thiazide-sensitive sodium-chloride cotransporter (NCC), which are normally inhibited by D5R [94]. Renal-selective depletion of Snx1 also leads to increased renal expression of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX) components, such as NOX1, NOX2, p47phox, resulting in oxidative stress [89,90]. Renal-selective Snx1 depletion also increases the renal expression of angiotensin II (Ang II) type 1 receptor (AT1R) and contributes to the increase in blood pressure which cannot be counteracted by the dysfunctional D5R [89,95,96]. These results indicate that SNX1-mediated regulation of D5R expression and function is needed for normal renal sodium handling and blood pressure.

Vascular SNXs and blood pressure

It cannot be overstated that abnormalities in the vasculature causes hypertension. In addition to the effects of SNXs in the kidney, SNXs regulate blood pressure by actions in the vasculature. We have reported that SNX1 and AT1R colocalize in the tunica media of the aorta and vascular smooth muscle cells (VSMCs) [97]. Snx1−/− mice, which as indicated earlier are hypertensive, have increased mesenteric arterial reactivity to Ang II, which is due to increased AT1R expression, without affecting the expression of the vasorelaxant AT2R. Snx1 knockdown with siRNA in VSMCs increases AT1R expression and decreases in the colocalization of AT1R and an endosome marker Rab5 [97]; the elevation in cytoplasmic Ca2+ level may explain the augmented VSMC reactivity in this situation [98,99]. Other studies in hRPTCs found that the elevation in AT1R protein may be a result of its reduced degradation in the proteasome pathway [96]. In Snx1−/− mice, the impaired function of D5R may be partly responsible for the increased expression of renal and vascular AT1R that is due to impaired SNX1-mediated AT1R sorting and degradation [89,90,97].

Vascular remodeling is both a cause and a consequence of hypertension [100]. Integrins, participating in the pathogenesis of vascular remodeling [101,102], are regulated by SNXs via different mechanisms. In endothelial cells, SNX9 determines the membrane levels of β1-integrins, an essential adhesion molecule; SNX9 depletion delays the β1-integrin cellular recycling and reduces the cell surface levels of β1-integrins, which are involved in the adhesion of VSMCs and endothelial cells [103,104]. SNX17 also enables β1-integrins to recycle to the cell surface and attenuates its transport to the lysosome for its decay by interacting with the β1-integrin’s tail [49]. SNX31, an SNX sharing with SNX17 their obligate phox domains, also modulates β1-integrin level at the membrane by binding to its tail [105]. Thus, SNXs, including SNX9, SNX17, and SNX31, can directly regulate the trafficking and expression of integrins, which may be involved in the modulation of vascular remodeling. However, more studies are needed to determine the role of SNX-regulated cellular localization of integrin in the pathogenesis of hypertension.

SNXs regulate vascular remodeling via other mechanisms. For example, SNX16 is also involved in the development of other types of hypertension, including pre-eclampsia [106]. Inflammation and immune mechanisms are involved in the pathogenesis of pre-eclampsia. SNX10 has been reported to regulate the inflammatory response. Snx10 deficient mice have increased basal and lipopolysaccharide-induced increase in anti-inflammatory M2 macrophages but decrease in M1 pro-inflammatory macrophages, which were verified by in vitro studies [107]. However, the role of different monocyte/macrophage populations in the pathogenesis of hypertension is complicated [108,109]. Thus, the M1 phenotype may predominate early while the M2 phenotype may predominate late in the disease process; the increase in M2 macrophages may then lower the blood pressure level [109]. SNX10 also modulates diet-induced atherogenesis by the Lyn-dependent transcription factor EB pathway [110]. Compared with wild-type (WT) mice, Snx10−/− mice have attenuated atherosclerotic plaque progression, as well as reduced foam cell amount [111]. In addition, SNX17 interacts with an adhesion protein P-selectin via its FERM domain, leading to the acceleration of cargo internalization and inhibition of the transport of P-selectin to lysosomes for its decay [112114]. However, the roles of SNX10 and SNX17 on blood pressure regulation have not been determined.

Regulation of SNXs on cellular sodium and water handling

As aforementioned, essential hypertension may be initiated by an increase in basal renal tubular sodium reabsorption but an appropriate decrease in renal tubular sodium reabsorption and increase in sodium excretion does not occur with the increase in blood pressure [63,115,116]. SNXs can directly affect cellular renal sodium handling. For example, SNX4 is required for the trafficking of the epithelial sodium channel (ENaC), which is involved in sodium reabsorption in the late distal convoluted tubule, connecting tubule, and collecting duct of the kidney [117120]. SNX4-mediated trafficking of ENaC may involve the kidney and brain protein, KIBRA [121]. Depletion of Snx27, a retromer-associated protein, does not alter ENaC activity [121]. However, SNX27 can directly interact with the C-terminus of NHE3 to regulate its trafficking to the renal brush border membrane [42]. SLC26A3, aka down-regulated adenoma protein (DRA) and chloride anion exchanger, with NHE3 mediate intestinal sodium absorption [122]. SNX27 regulates the activity of DRA by accelerating its recycling, but not its expression, to the membrane in intestinal cells [123].

There are other SNXs participating in the modulation of sodium and water handling. SNX11 facilitates the movement of transient receptor potential cation channel subfamily V member 3 (TRPV3) to lysosomes for its degradation [46]. SNX3 increases the cell surface expression of ENaC in HEK-293 cells and is required for the increase in ENaC cell surface expression mediated by vasopressin in mouse renal cortical collecting duct cells [124]. There is a co-localization of SNX27 and the water channel aquaporin 2 protein (AQP2) in the kidneys of rats. Further studies showed that in the rat renal collecting duct, the autophagy-lysosomal degradation of AQP2 is prevented by SNX27 [125]. Current studies have shown that SNXs, including SNX3, SNX4, SNX11, SNX27, can directly regulate the expression and/or activity of cellular sodium and water handling proteins by modulating their trafficking. However, the role, if any, of these SNXs in the regulation of blood pressure needs further studies.

SNXs can also indirectly regulate renal sodium handling. As discussed above, the depletion of renal Snx1 or Snx19 leads to increased activity of sodium pump, exchanger, and cotransporters by decreasing the D5R or D1R function [80,89,90].

SNXs and insulin resistance

Insulin resistance, a major metabolic risk factor, is involved in the pathogenesis of salt-induced hypertension [126]. Insulin production is regulated by SNXs, such as SNX1, SNX19, and SNX27. Both the amount of dense core vesicles (DCVs) and insulin production in β cells are reduced after Snx19 silencing, which are reversed with the re-expression of Snx19. These may be associated with the lysosome pathway because Snx19 silencing, in a mouse pancreatic β-cell line, increases the number of lysosomes, as well as the activity of cathepsin D, a lysosome enzyme [127]. SNXs also indirectly regulate insulin secretion. For example, SNX19 interacts with insulin antigen 2, part of DCVs that regulate insulin production, in part, by initiating the pre-apoptotic state of pancreatic β cells [128,129]. In mouse pancreatic β-cells, incretin-mediated insulin production is also increased after SNX27 or SNX1 silencing [130].

SNXs, including SNX1, SNX2, SNX4, SNX5, SNX9 and SNX27, can also regulate insulin-mediated signaling by regulating insulin receptor expression and its downstream signaling. SNX5, similar to SNX19, also regulates circulating insulin levels. The kidney is the main organ involved in the elimination of circulating insulin [131,132]. Our studies, and those of others, found that renal SNX5 positively regulates the expression and function of insulin-degrading enzyme (IDE), which is thought to be responsible for insulin clearance [133,134]. Co-localization of SNX5 and IDE is observed in RPTs of humans and mice, and at the plasma membrane and perinuclear area of hRPTCs, which are increased after insulin activation [134]. Depletion of renal Snx5 decreases IDE expression and activity, impairs insulin excretion from the urine, and responses to insulin and glucose, leading to the occurrence of insulin resistance. The levels of both SNX5 and IDE are also reduced in RPTCs from SHRs and hypertensive subjects [134].

Our previous studies also found that the expression of the insulin receptor is regulated by SNX5. There is a co-localization of SNX5 and insulin receptor throughout the RPTs of the human kidney, but their co-localization is only observed in brush borders and apical membranes of rat RPTs [59]. Snx5 depletion in hRPTCs reduces the amounts of insulin receptor and phosphorylated IRS-1 and PKB [59], the downstream signaling molecules of the insulin receptor [135,136]. Immuno-coprecipitation studies also showed that other SNXs, including SNX1, SNX2, and SNX4, can interact with the insulin receptor [137,138]. SNX9 is also involved in insulin action by stimulating insulin-stimulated glucose transport [139]. Proteomic analysis of SNX27 and retromer cargo specificity found that the glucose transporter, glucose transporter 1 (GLUT1), a critical signaling molecule of insulin, interacts with SNX27 and requires SNX27-retromer to prevent its decay in the lysosomes and preserve plasma cell membrane levels [50,140]. Glucose transporter 4 (GLUT4) is also modulated by SNXs, such as SNX27 and SNX9. In 3T3L1 adipocytes, Snx27 silencing with Snx27-shRNA decreases the expression of GLUT4 and blocking SNX9 with its antibody also inhibits insulin-mediated translocation of GLUT4 [139,141].

SNXs and hypertension

The deficiency of specific SNXs leads to the hypertensive phenotype (Table 2). Both systolic and diastolic blood pressures are higher in anesthetized Snx1−/− mice than WT mice [90]. Moreover, both nocturnal and diurnal systolic blood pressures are also elevated in conscious Snx1 knockout mice. The elevated blood pressure in Snx1−/− mice is not salt-sensitive because there are no differences of blood pressure in Snx1−/− mice fed with diets containing varying salt content [90]. The silencing of renal Snx1 also increases blood pressure in different strains of mice, e.g., C57Bl.6J and 129S1/SvlmJ mice [89]. The increased blood pressures in Snx1−/− mice or renal Snx1-silenced mice are accompanied by impaired D5R-dependent sodium excretion and increased AT1R-mediated reactivity of mesenteric artery [89,90]. Our previous studies also showed that silencing of renal Snx5 elevates the already increased systolic blood pressure in SHRs, accompanied by a decrease in 24-hour sodium excretion [77]. Moreover, knockdown of renal Snx5, by reducing the amount of IDE in RPTs, augments the blood levels of both insulin and glucose, leading to insulin resistance and hypertension [134]. We also found that the silencing of renal Snx19 elevates the blood pressure in mice fed with normal salt diet [80].

Table 2.

Summary of the role of SNX isoforms in hypertension

SNX isoform SNX variants or modifications Effects of SNX Modification on the regulation of blood pressure or related function SNX expression and function in human hypertension References
SNX1 Renal-selective Snx1 silencing (siRNA) Increases systolic and diastolic blood pressures, in salt-sensitive C57Bl/6J and salt-resistant BALB/cJ mice and impairs the natriuretic response to fenoldopam (D1R and D5R agonist) in salt-resistant BALB/cJ mice;
Increases renal AT1R protein expression in C57Bl/6J and BALB/cJ mice
Snx1 silencing (siRNA) in hRPTCs impairs D5R trafficking and function (stimulation of cAMP production and inhibition of sodium transport).
Decreased mRNA and protein expressions of SNX1, impaired SNX1-mediated D5R endocytosis, and increased ROS in RPTCs from hypertensive Euro-American males, relative to normotensive males
[89,90]
Global Snx1 knockout Increases systolic and diastolic blood pressures and impairs the natriuretic response to fenoldopam in 129S1/SvlmJ mice;
Increases AT1R protein expression in the kidney and aorta and reactivity of mesenteric artery to Ang II
Snx1 variants Not studied in rodents, as related to BP Snx1 variants, rs11635627, rs11854249 and rs12591927, are associated with impaired ability of HCTZ to decrease systolic blood pressure in African-Americans [90]
SNX5 Renal-selective Snx5 silencing (siRNA) Increases further the increased systolic blood pressure and decreased sodium excretion in SHRs;
Decreases renal IDE protein expression and urinary insulin excretion, and causes insulin resistance in C57Bl/6J mice;
Increases non-fasting serum insulin and glucose levels in normotensive WKY rats
Decreased SNX5 protein expression in RPTCs from hypertensive Euro-American males, relative to normotensive subjects [77,134]
SNX9 Snx9 variant Not studied in rodents, as related to BP Snx9 variant rs2364349 is associated with the heart rate lowering effect of β-blockers [150]
SNX10 Myeloid cell-specific SNX10 deficiency Snx10 depletion polarizes macrophages towards the anti-inflammatory M2 phenotype None [110,111]
SNX19 Renal-selective Snx19 silencing (siRNA) Increases the systolic blood pressure and decreases renal D1R expression of C57BL/6J mice fed a normal salt diet None [80]

Abbreviations: AT1R, angiotensin II type 1 receptor; β-blockers, β-adrenergic receptor blockers; BP, blood pressure; D1R, dopamine D1 receptor; D5R, dopamine D5 receptor; HCTZ, hydrochlorothiazide; IDE, insulin-degrading enzyme; ROS, reactive oxygen species; SHRs, spontaneously hypertensive rats; WKY, Wistar-Kyoto.

The renal expression of several SNXs is decreased in hypertensive states. Compared with normotensive subjects, the SNX5 protein expression is lower in RPTCs from hypertensive than normotensive subjects. This is consistent with the results in hypertensive animals; SNX5 expression is also lesser in the RPTCs and kidneys from SHRs than Wistar-Kyoto (WKY) rats [134]. Decreased renal SNX5 expression in SHRs may be a reason for their impaired ability to excrete a salt load. Indeed, silencing of renal Snx5 decreases sodium excretion and further elevates the already increased blood pressure of SHRs [77]. This is associated with impaired renal tubule cellular recycling of D1R, increased levels of phosphorylated D1R, and impaired D1R function, for example, impaired D1R-mediated increase in cAMP production [77]. We and others have reported that the renal D1R is important in the natriuresis with salt loading [5,142,143]. We also found that both mRNA and protein expressions of renal SNX1 are lower in hypertensive than normotensive Euro-American males [90]. The decreased amounts of renal SNXs may be a cause in the impaired dopamine receptor-mediated renal sodium handling in hypertension. The decreased levels of SNX1 in RPTCs from hypertensive humans blunts D5R trafficking, which leads to impaired renal D5R-mediated cAMP production and suppression of Na+-K+-ATPase activity. Silencing of renal Snx1 in mice increases blood pressure that is related to the impairment of a D1R and D5R agonist fenoldopam-mediated natriuretic effect [90], similar to that found with Snx5 silencing [77].

The Snx5 variant, rs6045116, and the five Snx19 variants, rs3751037, rs3190345, rs681982, rs4414223, rs2298566, are not associated with hypertension, regardless of salt sensitivity status in Americans of European ancestry [144]. However, there are a few studies showing the association between SNXs and hypertension or metabolic syndrome in humans. A GWAS showed the association of Snx19 and proliferation and apoptosis of pancreatic β-cells, which are involved in insulin production [145]. In addition, some Snx1 variants are associated with the response to antihypertensive drugs (see below).

SNXs and antihypertensive treatment

Methods of targeting SNXs

Targeting of SNXs, using several methods, has led to the identification of their roles in different diseases. Gene deletion or depletion, via siRNA or shRNA, and gene insertion/overexpression are widely used in the study of the association of diseases and genes, including SNXs. For example, Snx10 deficiency decreases foam cell abundance, alleviates atherosclerotic plaque progression, and reduces inflammation and pathological damage induced by dextran sulfate sodium [111,146]. By contrast, gene overexpression via plasmid or adenovirus transfection is used to enhance SNXs-mediated physiological effects. For example, SNX8 overexpression in human liver cells markedly blocks hepatocyte lipid deposition [147]. SNX1 gene-rescue in hRPTCs from hypertensive subjects restores the impaired D5R-mediated cAMP production and inhibition of renal sodium transport [90]. In addition, some novel methods, including nanoparticle-encapsulated Snx-shRNA plasmids with polylactide-polyglycolide or small-molecule inhibitor targeting of SNXs, have also been proved to be effective in altering gene expression [146,148]. However, it should be noted that while the biochemical role of SNXs appears largely conserved, the phenotypic consequences of loss of function may vary among species [149]. Thus, it is probably important to target the pathophysiological functions of aberrant SNXs in cells from hypertensive subjects, and not speculate or modify SNXs based entirely on the results of animal experimentations. Nevertheless, the associations between single nucleotide polymorphisms of SNXs and development of hypertension, as well as effects of antihypertensive treatment in several ethnic groups, have been reported (vide infra) [90,150].

SNX single nucleotide polymorphisms and antihypertensive effects

The potential role of SNXs in the treatment of hypertension has been demonstrated. Shahin et al. used GWAS to search genetic variants associated with heart rate during treatment with β-adrenergic receptor blockers (β-blockers) in Americans of European and African ancestry. They found that the Snx9 variant rs2364349 was associated with a decrease in heart rate following treatment with β-blockers. The decrease in heart rate was greater in subjects carrying Snx9 rs2364349 G/G and A/G genotypes than non-carriers [150].

We have also reported the association of SNX1 variants and response to antihypertensive drugs. We found twelve SNX1 variants in subjects with African- American ancestry and ten in subjects with European-American ancestry. In African-Americans, the SNX1 variants rs11635627and rs11854249 were associated with a decrease in systolic blood pressure with treatment with the diuretic hydrochlorothiazide [90]. We also found a trend of a decline in blood pressure with hydrochlorothiazide treatment in hypertensive patients with the SNX1 variant allele rs12591927 [90]. These data indicate that the SNX genotype may be predictive of the effect of anti-hypertensive treatment, a case of pharmacogenomics.

SNXs as potential antihypertensive targets

As discussed above, SNXs, such as SNX5 and SNX19, play roles in the development of hypertension in animal models. SNX27 may be a potential target of antihypertensive drugs because the recycling of β2-adrenergic receptors back to the membrane is blunted after Snx27 silencing [151]. β-adrenergic blockers are used in the treatment of hypertension [152]. Other SNXs, such as SNX3, SNX10, SNX13 and SNX17, reported to be associated with cardiovascular diseases, may also be antihypertensive targets [14,110,153155]. Indeed, SNXs are associated with risk factors, e.g., inflammation, involved the development of hypertension. For example, impaired SNX9 expression has been reported in different animal models of chronic inflammation, including chronic infection (Leishmania donovani), and autoimmune disease (rheumatoid arthritis) [156]. Hypertension is associated with chronic inflammation [157,158]. Thus, SNXs associated with chronic inflammation may be also involved in the pathogenesis of hypertension. However, the direct correlation between the above-mentioned SNXs and hypertension or antihypertensive effects needs to be clarified in the future.

SNXs and anti-cancer drug-induced hypertension

Angiogenesis, the formation of new blood vessels, is involved in the development and progression of many diseases, including cancers [159]. Some anti-cancer drugs, including monoclonal antibodies that inhibit angiogenesis or proliferation, can increase blood pressure [160,161]. Thus, it is important to find new strategies to decrease the anti-angiogenesis drug-induced hypertension with anti-cancer therapy. We suggest that targeting SNXs may be a potential strategy to reduce the cancer-associated hypertension for three reasons:

  1. SNXs are associated with carcinoma progression. For example, SNX1 serves as a tumor suppressor and potential prognostic marker in colorectal cancer and gastric cancer [162,163]. Snx1 depletion in colon cancer cells increases cell proliferation and decreases apoptosis, which would promote colon tumorigenesis [164]. SNX5 expression is increased in hepatocellular carcinoma, which promotes vascular invasion and intrahepatic metastasis [165]. Snx5 depletion decreases cell proliferation, migration, and invasion [165].

  2. SNXs regulate the expression and trafficking of some growth factor receptors, which are involved in the regulation of both cancerogenesis and blood pressure. For example, EGFR, a transmembrane glycoprotein belonging to the ERBB family of tyrosine kinase receptors, plays a vital role in signal transduction and oncogenesis [166,167]. Abnormal endocytosis and trafficking of EGFR contribute to aberrant EGFR signaling in cancer [168]. Overexpression of Snx1 promotes EGFR degradation in lysosomes and decreases the amount of EGFR on the cell surface [44,45]. SNX2, which has a redundant function with SNX1, can also participate in the lysosomal sorting of EGFRs [169,170]. SNX5 interacts with EGFR in hepatocellular carcinoma cells; SNX5 promotes cell proliferation, migration, and invasion, via the activation of EGFR by blocking EGF-mediated EGFR internalization, which is reversed by the knockdown of EGFR [165]. Other SNXs, including SNX3, SNX6 and SNX16, directly regulate the intracellular trafficking and sorting of EGFRs [171173].

  3. SNXs sensitize cancer cells to anti-angiogenesis drugs by regulating the activity and trafficking of EGFRs. For example, gefitinib, a specific inhibitor of EGFR tyrosine kinase, is known to repress the activation of EGFR signaling required for cell growth in non-small cell lung cancer (NSCLC) cell lines. In the gefitinib-sensitive NSCLC cell line PC9, there is efficient endocytosis of phosphorylated EGFR which leads to its degradation. However, in gefitinib-resistant NSCLC cell lines, the internalized phosphorylated EGFR accumulates in early endosomes, not in late endosomes/lysosomes for degradation; this process can be normalized by Snx1 depletion [174176]. However, other studies have reported that SNX1 actually enhances the degradation of EGFR [44,177]. Snx2 silencing in human lung cancer cells enhances the sensitivity to gefitinib and erlotinib, another anti-angiogenic drug that targets EGFR [178]. Snx5 overexpression promotes while its silencing inhibits hepatocellular carcinoma cell proliferation, migration, and invasion that are related to activation of EGFR [165]. As is the case with Snx2 silencing, Snx5 silencing increases the sensitivity to EGFR inhibitors, e.g., erlotinib and sorafenib [165]. These findings suggest that targeting SNXs may be useful in overcoming drug resistance to EGFR-targeted drugs in cancer cells. Moreover, the SNXs-mediated increase in sensitivity of cancer cells to anti-angiogenesis drugs may lower the dosage of these anti-cancer drugs and reduce the risk of drug-induced high blood pressure. However, there is lack of direct evidence to show the effect of targeting SNXs on the regulation of anti-angiogenesis drug-induced hypertension, which remains to be determined.

Conclusion

In summary, SNXs exert their biological functions by regulating cellular cargo sorting and trafficking, maintaining intracellular protein homeostasis, and participating in intracellular signaling cascade. Increasing pieces of evidence demonstrate that SNXs exert important functions to maintain a normal blood pressure, via different mechanisms (Figure 1). Dysfunction of SNXs in the cardiovascular system and kidney participate in the development of hypertension. Moreover, genetic studies have also shown an association of SNXs gene polymorphisms and responses to anti-hypertensive drugs. However, there remain some important questions in the role of SNXs in blood pressure regulation. For example: 1) the role of SNXs in the development and treatment of hypertension needs to be studied further; 2) most studies focus on the regulation of SNXs on the trafficking of intracellular cargos. There are few studies exploring how the expression and function of SNXs are regulated [179]; 3) in addition to the kidney and vasculature, the role of SNXs in other blood pressure-related organs or target organs in hypertension needs to be determined; and 4) further studies on the crystal structure of SNX domains, SNX-mediated membrane remodeling, and interaction with their targets may be helpful in understanding SNX regulatory mechanisms in hypertension and anti-hypertensive treatment [180,181]. Such studies will improve our understanding of the role of SNXs in the regulation of blood pressure and provide novel strategies for the treatment of hypertension.

Figure 1. Regulation of blood pressure by SNXs.

Figure 1.

SNXs regulate blood pressure via different mechanisms. In the artery, SNX1 negatively regulates AT1R-mediated vasoconstriction by increasing AT1R-mediated proteasomal degradation; SNXs9/17/31 promote the plasma membrane recycling of β1-integrin; SNX10 promotes the polarization of macrophages towards the MI pro-inflammatory phenotype. In the kidney, SNXs1/5/19 promote D1R- and D5R-induced natriuresis. SNXs regulate cellular sodium and water transport: SNXs3/4 regulate the trafficking of ENaC, increasing its membrane expression and therefore activity; SNX27 increases the activity of NHE3, DRA (aka SLC26A3), and AQP2 by increasing their recycling to the plasma membrane; and SNX11 facilitates the movement of TRPV3 to lysosomes, increasing its degradation. SNXs regulate insulin production and response: SNXs1/19/27 increase insulin secretion; SNXs1/2/4 co-immunoprecipitate with the insulin receptor; and SNX5 negatively regulates the expression of the renal insulin receptor and insulin clearance by increasing the expression and function of IDE. SNXs9/27 increase the expression or plasma membrane translocation of GLUT1/4. Dysfunctions of SNXs participate in the pathogenesis and pharmacogenomics of hypertension. The response to antihypertensive drugs is influenced by variants of some SNXs. For example, the Snx9 variant rs2364349 is associated with the decrease in heart rate following treatment with β-blockers; the decrease in systolic blood pressure, in response to the diuretic HCTZ, is associated with the SNX1 variants rs11635627and rs11854249; Snx2 silencing in human lung cancer cells enhances the sensitivity to the tyrosine kinase inhibitors, gefitinib and erlotinib, but may also cause hypertension.

Abbreviation: AT1R: angiotensin II (Ang II) type 1 receptor; AQP2: aquaporin-2 protein; β-blockers: β-adrenergic receptor blockers; D1R: dopamine D1 receptor; D5R: dopamine D5 receptor; DRA, down-regulated adenoma protein (also known as chloride ion exchanger and SLC26A3, solute carrier family 26 member 3); ENaC: epithelial sodium channel; GLUT1: glucose transporter type 1; GLUT4: glucose transporter type 4; HCTZ: hydrochlorothiazide; IDE: insulin-degrading enzyme; NHE3, sodium-hydrogen exchanger type 3; SNX: sorting nexin.

Acknowledgements

The work was funded by grants from the National Natural Science Foundation of China (81770425, 82070442), Key Program of The Third Affiliated Hospital of Chongqing Medical University (KY19024), and the National Institutes of Health (5R01DK039308, 5P01HL074940, and R01DK119652).

Abbreviations

Ang II

angiotensin II

AT1R

angiotensin II type 1 receptor

AQP2

aquaporin 2

ATG

autophagy-related gene

BAR

Bin/Amphiphysin/Rvs

β-blockers

β-adrenergic receptor blockers

cAMP

cyclic adenosine monophosphate

DCVs

dense core vesicles

D1R

dopamine D1 receptor

D5R

dopamine D5 receptor

DRA

down-regulated adenoma protein (also known as chloride ion exchanger and SLC26A3, solute carrier family 26 member 3)

EGFR

epidermal growth factor receptor

ENaC

epithelial sodium channel

FERM

protein 4.1/ezrin/radixin/moesin

GLUT1

glucose transporter type 1

GLUT4

glucose transporter type 4

GPCRs

G protein-coupled receptors

HCTZ

hydrochlorothiazide

hRPTCs

human renal proximal tubule cells

IDE

insulin-degrading enzyme

IR

insulin receptor

IRS-1

insulin receptor substrate-1

NADPH

nicotinamide adenine dinucleotide phosphate

NCC

thiazide-sensitive sodium-chloride cotransporter

NHE3

sodium-hydrogen exchanger type 3

NOX

NADPH oxidase

NSCLC

non-small cell lung cancer

PX

phagocyte oxidase (phox) homology domain

PDZ

postsynaptic density 95/discs large/zonula occludens

PI3P

phosphatidylinositol-3-phosphate

PXA

PX-associated domain A

PXC

PX-associated domain C

RGS

regulator of G-protein signaling

RPTs

renal proximal tubules

SHRs

spontaneously hypertensive rats

SNX

sorting nexin

TGF-β

transforming growth factor β

TRPV3

transient receptor potential cation channel subfamily V member 3

VSMCs

vascular smooth muscle cells

WKY

Wistar-Kyoto

WT

wild-type

Footnotes

Conflicts of interest

The authors declare that they have no competing interests.

References

  • 1.Virani SS, Alonso A, Benjamin EJ, Bittencourt MS, Callaway CW, Carson AP, Chamberlain AM, Chang AR, Cheng S, Delling FN et al. (2020) Heart disease and stroke statistics-2020 update: a report from the American Heart Association. Circulation 141, e139–e596. [DOI] [PubMed] [Google Scholar]
  • 2.Kearney PM, Whelton M, Reynolds K, Muntner P, Whelton PK & He J (2005) Global burden of hypertension: analysis of worldwide data. Lancet 365, 217–223. [DOI] [PubMed] [Google Scholar]
  • 3.Mills KT, Bundy JD, Kelly TN, Reed JE, Kearney PM, Reynolds K, Chen J & He J (2016) Global disparities of hypertension prevalence and control: a systematic analysis of population-based studies from 90 countries. Circulation 134, 441–450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Gheorghe A, Griffiths U, Murphy A, Legido-Quigley H, Lamptey P & Perel P (2018) The economic burden of cardiovascular disease and hypertension in low- and middle-income countries: a systematic review. BMC Public Health 18, 975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Qaddumi WN & Jose PA (2021) The role of the renal dopaminergic system and oxidative stress in the pathogenesis of hypertension. Biomedicines 9, 139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Vamvakis A, Gkaliagkousi E, Lazaridis A, Grammatikopoulou MG, Triantafyllou A, Nikolaidou B, Koletsos N, Anyfanti P, Tzimos C, Zebekakis P et al. (2020) Impact of intensive lifestyle treatment (diet plus exercise) on endothelial and vascular function, arterial stiffness and blood pressure in stage 1 hypertension: results of the HINTreat randomized controlled trial. Nutrients 12, 1326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Hu H, Li H, Huang X, Bao H, Song Y, Wang B, Liu C, Xu R, Liu L, Wang X et al. (2020) Association of self-reported sleep duration and quality with BaPWV levels in hypertensive patients. Hypertens Res 43, 1392–1402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Seidel E & Scholl UI (2017) Genetic mechanisms of human hypertension and their implications for blood pressure physiology. Physiol Genomics 49, 630–652. [DOI] [PubMed] [Google Scholar]
  • 9.Liang M (2018) Epigenetic mechanisms and hypertension. Hypertension 72, 1244–1254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Cullen PJ & Korswagen HC (2011) Sorting nexins provide diversity for retromer-dependent trafficking events. Nat Cell Biol 14, 29–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Chen KE, Healy MD & Collins BM (2019) Towards a molecular understanding of endosomal trafficking by Retromer and Retriever. Traffic 20, 465–478. [DOI] [PubMed] [Google Scholar]
  • 12.Chandra M, Kendall AK & Jackson LP (2020) Unveiling the cryo-EM structure of retromer. Biochem Soc Trans 48, 2261–2272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Yang J, Villar VAM, Rozyyev S, Jose PA & Zeng C (2019). The emerging role of sorting nexins in cardiovascular diseases. Clin Sci (Lond) 133, 723–737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Chandra M, Kendall AK & Jackson LP (2021) Toward understanding the molecular role of SNX27/retromer in human health and disease. Front Cell Dev Biol. 2021, 9, 642378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Wang X, Zhao Y, Zhang X, Badie H, Zhou Y, Mu Y, Loo LS, Cai L, Thompson RC, Yang B et al. (2013) Loss of sorting nexin 27 contributes to excitatory synaptic dysfunction by modulating glutamate receptor recycling in Down’s syndrome. Nat Med 19, 473–480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Cullen PJ (2008) Endosomal sorting and signalling: an emerging role for sorting nexins. Nat Rev Mol Cell Biol 9, 574–582. [DOI] [PubMed] [Google Scholar]
  • 17.Yang L, Tan W, Yang X, You Y, Wang J, Wen G & Zhong J (2020) Sorting nexins: a novel promising therapy target for cancerous/neoplastic diseases. J Cell Physiol. doi: 10.1002/jcp.30093. [DOI] [PubMed] [Google Scholar]
  • 18.Chandra M & Collins BM (2019) The phox homology (PX) domain. Adv Exp Med Biol 1111, 1–17. [DOI] [PubMed] [Google Scholar]
  • 19.Chandra M, Chin YK, Mas C, Feathers JR, Paul B, Datta S, Chen KE, Jia X, Yang Z, Norwood SJ et al. (2019) Classification of the human phox homology (PX) domains based on their phosphoinositide binding specificities. Nat Commun 10, 1528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.van Weering JR, Verkade P & Cullen PJ (2010) SNX-BAR proteins in phosphoinositide-mediated, tubular-based endosomal sorting. Semin Cell Dev Biol. 21, 371–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Ghai R, Mobli M, Norwood SJ, Bugarcic A, Teasdale RD, King GF & Collins BM (2011) Phox homology band 4.1/ezrin/radixin/moesin-like proteins function as molecular scaffolds that interact with cargo receptors and Ras GTPases. Proc Natl Acad Sci U S A 108, 7763–7768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Yong X, Zhao L, Hu W, Sun Q, Ham H, Liu Z, Ren J, Zhang Z, Zhou Y, Yang Q, et al. (2021) SNX27-FERM-SNX1 complex structure rationalizes divergent trafficking pathways by SNX17 and SNX27. Proc Natl Acad Sci U S A 118, e2105510118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.DuChez BJ, Hueschen CL, Zimmerman SP, Baumer Y, Wincovitch S & Playford MP (2019) Characterization of the interaction between beta-catenin and sorting nexin 27: contribution of the type I PDZ-binding motif to Wnt signaling. Biosci Rep 39, BSR20191692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Zheng B, Ma YC, Ostrom RS, Lavoie C, Gill GN, Insel PA, Huang XY & Farquhar MG (2001) RGS-PX1, a GAP for GalphaS and sorting nexin in vesicular trafficking. Science 294, 1939–1942. [DOI] [PubMed] [Google Scholar]
  • 25.Zheng B, Tang T, Tang N, Kudlicka K, Ohtsubo K, Ma P, Marth JD, Farquhar MG & Lehtonen E (2006) Essential role of RGS-PX1/sorting nexin 13 in mouse development and regulation of endocytosis dynamics. Proc Natl Acad Sci U S A 103, 16776–16781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Rodríguez-Fraticelli AE, Galvez-Santisteban M & Martin-Belmonte F (2013) KIF16B delivers for transcytosis. EMBO J 32, 2093–2095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Danson C, Brown E, Hemmings OJ, McGough IJ, Yarwood S, Heesom KJ, Carlton JG, Martin-Serrano J, May MT, Verkade P et al. (2013) SNX15 links clathrin endocytosis to the PtdIns3P early endosome independently of the APPL1 endosome. J Cell Sci 126, 4885–4899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Lucas M, Gershlick DC, Vidaurrazaga A, Rojas AL, Bonifacino JS & Hierro A (2016) Structural mechanism for cargo recognition by the retromer complex. Cell 167, 1623–1635.e14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Macías-Calvio V, Fuentealba LM & Marzolo MP (2021) An update on cellular and molecular determinants of Parkinson’s disease with emphasis on the role of the retromer complex. J Neurosci Res 99, 163–179. [DOI] [PubMed] [Google Scholar]
  • 30.McNally KE & Cullen PJ (2018) Endosomal retrieval of cargo: retromer is not alone. Trends Cell Biol 28, 807–822. [DOI] [PubMed] [Google Scholar]
  • 31.Han J, Goldstein LA, Hou W, Watkins SC & Rabinowich H (2020) Involvement of CASP9 (caspase 9) in IGF2R/CI-MPR endosomal transport. Autophagy 17, 1393–1409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.McGarvey JC, Xiao K, Bowman SL, Mamonova T, Zhang Q, Bisello A, Sneddon WB, Ardura JA, Jean-Alphonse F, Vilardaga JP et al. (2016) Actin-sorting nexin 27 (SNX27)-retromer complex mediates rapid parathyroid hormone receptor recycling. J Biol Chem 291, 10986–1002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Feng S, Streets AJ, Nesin V, Tran U, Nie H, Onopiuk M, Wessely O, Tsiokas L, & Ong ACM (2017) The sorting nexin 3 retromer pathway regulates the cell surface localization and activity of a Wnt-activated polycystin channel complex. J Am Soc Nephrol 28, 2973–2984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Tian Y, Kang Q, Shi X, Wang Y, Zhang N, Ye H, Xu Q, Xu T, & Zhang R (2021) SNX-3 mediates retromer-independent tubular endosomal recycling by opposing EEA-1-facilitated trafficking. PLoS Genet 17, e1009607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Hanley SE & Cooper KF (2020) Sorting nexins in protein homeostasis. Cells 10, 17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Tu Y & Seaman MNJ (2021) Navigating the controversies of retromer-mediated endosomal protein sorting. Front Cell Dev Biol 9, 658741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Schmid SL (2019) A nostalgic look back 40 years after the discovery of receptor-mediated endocytosis. Mol Biol Cell 30, 1–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Sasaki Y, Hidaka T, Ueno T, Akiba-Takagi M, Oliva Trejo JA, Seki T, Nagai-Hosoe Y, Tanaka E, Horikoshi S et al. (2017) Sorting Nexin 9 facilitates podocin endocytosis in the injured podocyte. Sci Rep 7, 43921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Bendris N & Schmid SL (2017) Endocytosis, metastasis and beyond: multiple facets of SNX9. Trends Cell Biol 27, 189–200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Schöbel S, Neumann S, Hertweck M, Dislich B, Kuhn PH, Kremmer E, Seed B, Baumeister R, Haass C & Lichtenthaler SF (2008) A novel sorting nexin modulates endocytic trafficking and alpha-secretase cleavage of the amyloid precursor protein. J Biol Chem 283, 14257–14268. [DOI] [PubMed] [Google Scholar]
  • 41.Sharma R, Gulia R & Bhattacharyya S (2018) A critical role for sorting nexin 1 in the trafficking of metabotropic glutamate receptors. J Neurosci 38, 8605–8620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Singh V, Yang J, Cha B, Chen TE, Sarker R, Yin J, Avula LR, Tse M & Donowitz M (2015) Sorting nexin 27 regulates basal and stimulated brush border trafficking of NHE3. Mol Biol Cell 26, 2030–2043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Shen Z, Li Y, Fang Y, Lin M, Feng X, Li Z, Zhan Y, Liu Y, Mou T, Lan X, et al. (2020) SNX16 activates c-Myc signaling by inhibiting ubiquitin-mediated proteasomal degradation of eEF1A2 in colorectal cancer development. Mol Oncol 14, 387–406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Kurten RC, Cadena DL & Gill GN (1996) Enhanced degradation of EGF receptors by a sorting nexin, SNX1. Science 272, 1008–1010. [DOI] [PubMed] [Google Scholar]
  • 45.Chin LS, Raynor MC, Wei X, Chen HQ & Li L (2001) Hrs interacts with sorting nexin 1 and regulates degradation of epidermal growth factor receptor. J Biol Chem 276, 7069–7078. [DOI] [PubMed] [Google Scholar]
  • 46.Li C, Ma W, Yin S, Liang X, Shu X, Pei D, Egan TM, Huang J, Pan A & Li Z (2016) Sorting nexin 11 regulates lysosomal degradation of plasma membrane TRPV3. Traffic 17, 500–514. [DOI] [PubMed] [Google Scholar]
  • 47.Sun M, Han X, Chang F, Xu H, Colgan L & Liu Y (2020) Regulatory role of sorting nexin 5 in protein stability and vesicular targeting of vesicular acetylcholine transporter to synaptic vesicle-like vesicles in PC12 cells. J Biomed Res doi: 10.7555/JBR.34.20200095. Online ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Zhang S, Yang Z, Bao W, Liu L, You Y, Wang X, Shao L, Fu W, Kou X, Shen W et al. (2020) SNX10 (sorting nexin 10) inhibits colorectal cancer initiation and progression by controlling autophagic degradation of SRC. Autophagy 16, 735–749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Böttcher RT, Stremmel C, Meves A, Meyer H, Widmaier M, Tseng HY & Fässler R (2012) Sorting nexin 17 prevents lysosomal degradation of beta1 integrins by binding to the beta1-integrin tail. Nat Cell Biol 14, 584–592. [DOI] [PubMed] [Google Scholar]
  • 50.Steinberg F, Gallon M, Winfield M, Thomas EC, Bell AJ, Heesom KJ, Tavaré JM & Cullen PJ (2013) A global analysis of SNX27-retromer assembly and cargo specificity reveals a function in glucose and metal ion transport. Nat Cell Biol 15, 461–471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Shen AW, Fu LL, Lin L, Sun B, Song DX, Wang WT, Wang YH, Yin PR & Yu SQ (2020) SNX9 inhibits cell proliferation and Cyst development in autosomal dominant polycystic kidney disease via activation of the Hippo-YAP signaling pathway. Front Cell Dev Biol 8:811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Xu L, Yin W, Xia J, Peng M, Li S, Lin S, Pei D & Shu X (2012) An antiapoptotic role of sorting nexin 7 is required for liver development in zebrafish. Hepatology 55, 1985–1993. [DOI] [PubMed] [Google Scholar]
  • 53.Dong X, Yang Y, Zou Z, Zhao Y, Ci B, Zhong L, Bhave M, Wang L, Kuo YC, Zang X et al. (2021) Sorting nexin 5 mediates virus-induced autophagy and immunity. Nature 589, 456–461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Ravussin A, Brech A, Tooze SA & Stenmark H (2021) The phosphatidylinositol 3-phosphate-binding protein SNX4 controls ATG9A recycling and autophagy. J Cell Sci 134, jcs250670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Si W, Huang Z, Li X, Zhao L, Ji Y, Li H, Liu X, Ye S, Chen D, Liu H et al. (2021) Super-enhancer-driven Sorting Nexin 5 expression promotes dopaminergic neuronal ferroptosis in Parkinson’s disease models. Biochem Biophys Res Commun 567, 35–41. [DOI] [PubMed] [Google Scholar]
  • 56.Le Y, Zhang S, Ni J, You Y, Luo K, Yu Y & Shen X (2018) Sorting nexin 10 controls mTOR activation through regulating amino-acid metabolism in colorectal cancer. Cell Death Dis 9, 666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Wilkes MC, Repellin CE, Kang JH, Andrianifahanana M, Yin X & Leof EB (2015) Sorting nexin 9 differentiates ligand-activated Smad3 from Smad2 for nuclear import and transforming growth factor beta signaling. Mol Biol Cell 26, 3879–3891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Kang JH, Jung MY, Yin X, Andrianifahanana M, Hernandez DM & Leof EB (2017) Cell-penetrating peptides selectively targeting SMAD3 inhibit profibrotic TGF-beta signaling. J Clin Invest 127, 2541–2554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Li F, Yang J, Jones JE, Villar VA, Yu P, Armando I, Felder RA & Jose PA (2015) Sorting nexin 5 and dopamine d1 receptor regulate the expression of the insulin receptor in human renal proximal tubule cells. Endocrinology 156, 2211–2221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Saric A, Freeman SA, Williamson CD, Jarnik M, Guardia CM, Fernandopulle MS, Gershlick DC & Bonifacino JS (2021) SNX19 restricts endolysosome motility through contacts with the endoplasmic reticulum. Nat Commun 12, 4552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Datta S, Bowerman J, Hariri H, Ugrankar R, Eckert KM, Corley C, Vale G, McDonald JG & Henne WM (2020) Snx14 proximity labeling reveals a role in saturated fatty acid metabolism and ER homeostasis defective in SCAR20 disease. Proc Natl Acad Sci U S A 117, 33282–33294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Bryant D, Liu Y, Datta S, Hariri H, Seda M, Anderson G, Peskett E, Demetriou C, Sousa S, Jenkins D, et al. (2018) SNX14 mutations affect endoplasmic reticulum-associated neutral lipid metabolism in autosomal recessive spinocerebellar ataxia 20. Hum Mol Genet 27, 1927–1940. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Hall JE, do Carmo JM, da Silva AA, Wang Z & Hall ME (2019) Obesity, kidney dysfunction and hypertension: mechanistic links. Nat Rev Nephrol 15, 367–385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Yang N, Hong NJ & Garvin JL (2020) Dietary fructose enhances angiotensin II-stimulated Na(+) transport via activation of PKC-alpha in renal proximal tubules. Am J Physiol Renal Physiol 318, F1513–F1519 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Li XC, Zhu D, Chen X, Zheng X, Zhao C, Zhang J, Soleimani M, Rubera I, Tauc M, Zhou X et al. (2019) Proximal tubule-specific deletion of the NHE3 (Na(+)/H(+) Exchanger 3) in the kidney attenuates ang II (angiotensin II)-induced hypertension in mice. Hypertension 74, 526–535 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Gildea JJ, Xu P, Kemp BA, Carey RM, Jose PA & Felder RA (2019) The dopamine D(1) receptor and angiotensin II type-2 receptor are required for inhibition of sodium transport through a protein phosphatase 2A pathway. Hypertension 73, 1258–1265 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Banday AA & Lokhandwala MF (2020) Renal dopamine oxidation and inflammation in high salt fed rats. J Am Heart Assoc 9, e014977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Zhang MZ & Harris RC (2015) Antihypertensive mechanisms of intra-renal dopamine. Curr Opin Nephrol Hypertens 24, 117–122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Zeng C & Jose PA (2011) Dopamine receptors: important antihypertensive counterbalance against hypertensive factors. Hypertension 57, 11–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Bek MJ, Wang X, Asico LD, Jones JE, Zheng S, Li X, Eisner GM, Grandy DK, Carey RM, Soares-da-Silva P et al. (2006) Angiotensin-II type 1 receptor-mediated hypertension in D4 dopamine receptor-deficient mice. Hypertension 47, 288–295. [DOI] [PubMed] [Google Scholar]
  • 71.Zeng C, Liu Y, Wang Z, He D, Huang L, Yu P, Zheng S, Jones JE, Asico LD, Hopfer U et al. (2006) Activation of D3 dopamine receptor decreases angiotensin II type 1 receptor expression in rat renal proximal tubule cells. Circ Res 99, 494–500. [DOI] [PubMed] [Google Scholar]
  • 72.Yu P, Asico LD, Luo Y, Andrews P, Eisner GM, Hopfer U, Felder RA & Jose PA (2006) D1 dopamine receptor hyperphosphorylation in renal proximal tubules in hypertension. Kidney Int 70, 1072–1079. [DOI] [PubMed] [Google Scholar]
  • 73.Banday AA, Diaz AD & Lokhandwala M (2019) Kidney dopamine D(1)-like receptors and angiotensin 1–7 interaction inhibits renal Na(+) transporters. Am J Physiol Renal Physiol 317, F949–F956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Jose PA, Eisner GM, Drago J, Carey RM & Felder RA (1996) Dopamine receptor signaling defects in spontaneous hypertension. Am J Hypertens 9,400–405. [DOI] [PubMed] [Google Scholar]
  • 75.Albrecht FE, Drago J, Felder RA, Printz MP, Eisner GM, Robillard JE, Sibley DR, Westphal HJ & Jose P (1996) Role of the D1A dopamine receptor in the pathogenesis of genetic hypertension. J Clin Invest 97, 2283–2288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Zeng C, Armando I, Luo Y, Eisner GM, Felder RA & Jose PA (2008) Dysregulation of dopamine-dependent mechanisms as a determinant of hypertension: studies in dopamine receptor knockout mice. Am J Physiol Heart Circ Physiol 294, H551–H569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Villar VA, Armando I, Sanada H, Frazer LC, Russo CM, Notario PM, Lee H, Comisky L, Russell HA, Yang Y et al. (2013) Novel role of sorting nexin 5 in renal D(1) dopamine receptor trafficking and function: implications for hypertension. FASEB J 27, 1808–1819. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Guo Y & Jose PA (2011) C-terminal di-leucine motif of dopamine D1 receptor plays an important role in its plasma membrane trafficking. PLoS One 6, e29204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Lamey M, Thompson M, Varghese G, Chi H, Sawzdargo M, George SR & O’Dowd BF (2002) Distinct residues in the carboxyl tail mediate agonist-induced desensitization and internalization of the human dopamine D1 receptor. J Biol Chem 277, 9415–9421. [DOI] [PubMed] [Google Scholar]
  • 80.Tiu AC, Yang J, Asico LD, Konkalmatt P, Zheng X, Cuevas S, Wang X, Lee H, Mazhar M, Felder RA et al. (2020) Lipid rafts are required for effective renal D1 dopamine receptor function. FASEB J 34, 6999–7017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Martinez VJ, Asico LD, Jose PA & Tiu AC (2020) Lipid rafts and dopamine receptor signaling. Int J Mol Sci 21, 8909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Mohamed A, Robinson H, Erramouspe PJ & Hill MM (2018) Advances and challenges in understanding the role of the lipid raft proteome in human health. Expert Rev Proteomics 15, 1053–1063. [DOI] [PubMed] [Google Scholar]
  • 83.Free RB, Hazelwood LA, Spalding HN, Cabrera DM & Sibley DR (2007) Sorting nexin-25, a novel member of the dopamine receptor signalplex, upregulates D1 and D2 dopamine receptor expression in HEK293 cells. FASEB J 21, 568.9. [Google Scholar]
  • 84.Free RB, Namkung Y, Hazelwood LA & Sibley DR (2010) Sorting nexin-25 interacts with D1 and D2 dopamine receptors to regulate receptor expression and signaling. FASEB J 24, 771.8. [Google Scholar]
  • 85.Rao F, Wessel J, Wen G, Zhang L, Rana BK, Kennedy BP, Greenwood TA, Salem RM, Chen Y, Khandrika S, et al. (2007) Renal albumin excretion: twin studies identify influences of heredity, environment, and adrenergic pathway polymorphism. Hypertension 49, 1015–1031. [DOI] [PubMed] [Google Scholar]
  • 86.Pontremoli R, Viazzi F, Sofia A, Tomolillo C, Ruello N, Bezante GP, Del Sette M, Martinoli C, Lotti R, Sacchi G, et al. (1997) Microalbuminuria: a marker of cardiovascular risk and organ damage in essential hypertension. Kidney Int Suppl 63, S163–S165. [PubMed] [Google Scholar]
  • 87.Asico L, Zhang X, Jiang J, Cabrera D, Escano CS, Sibley DR, Wang X, Yang Y, Mannon R, Jones JE et al. (2011) Lack of renal dopamine D5 receptors promotes hypertension. J Am Soc Nephrol 22:82–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Sunahara RK, Guan HC, O’Dowd BF, Seeman P, Laurier LG, Ng G, George SR, Torchia J, Van Tol HH & Niznik HB (1991) Cloning of the gene for a human dopamine D5 receptor with higher affinity for dopamine than D1. Nature 350, 614–619. [DOI] [PubMed] [Google Scholar]
  • 89.Villar VA, Jones JE, Armando I, Asico LD, Escano CS Jr, Lee H, Wang X, Yang Y, Pascua-Crusan AM, Palmes-Saloma CP et al. (2013) Sorting nexin 1 loss results in D5 dopamine receptor dysfunction in human renal proximal tubule cells and hypertension in mice. J Biol Chem 288, 152–163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Yang J, Asico LD, Beitelshees AL, Feranil JB, Wang X, Jones JE, Armando I, Cuevas SG, Schwartz GL, Gums JG, et al. (2020) Sorting nexin 1 loss results in increased oxidative stress and hypertension. FASEB J 34, 7941–7957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Griffin CT, Trejo J & Magnuson T (2005) Genetic evidence for a mammalian retromer complex containing sorting nexins 1 and 2. Proc Natl Acad Sci U S A 102, 15173–15177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Nisar S, Kelly E, Cullen PJ & Mundell SJ (2010) Regulation of P2Y1 receptor traffic by sorting nexin 1 is retromer independent. Traffic 11, 508–519. [DOI] [PubMed] [Google Scholar]
  • 93.Heydorn A, Søndergaard BP, Hadrup N, Holst B, Haft CR & Schwartz TW (2004) Distinct in vitro interaction pattern of dopamine receptor subtypes with adaptor proteins involved in post-endocytotic receptor targeting. FEBS Lett 556, 276–280. [DOI] [PubMed] [Google Scholar]
  • 94.Wang X, Luo Y, Escano CS, Yang Z, Asico L, Li H, Jones JE, Armando I, Lu Q, Sibley DR et al. (2010) Upregulation of renal sodium transporters in D5 dopamine receptor-deficient mice. Hypertension 55, 1431–1437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Li H, Armando I, Yu P, Escano C, Mueller SC, Asico L, Pascua A, Lu Q, Wang X, Villar VA et al. (2008) Dopamine 5 receptor mediates Ang II type 1 receptor degradation via a ubiquitin-proteasome pathway in mice and human cells. J Clin Invest 118, 2180–2189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Gildea JJ, Wang X, Jose PA & Felder RA (2008) Differential D1 and D5 receptor regulation and degradation of the angiotensin type 1 receptor. Hypertension 51, 360–366. [DOI] [PubMed] [Google Scholar]
  • 97.Liu C, Li X, Fu J, Chen K, Liao Q, Wang J, Chen C, Luo H, Jose PA, Yang Y, et al. (2021) Increased AT1 receptor expression mediates vasoconstriction leading to hypertension in Snx1−/− mice. Hypertens Res 44, 906–917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Touyz RM, Alves-Lopes R, Rios FJ, Camargo LL, Anagnostopoulou A, Arner A & Montezano AC (2018) Vascular smooth muscle contraction in hypertension. Cardiovasc Res 114, 529–539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Chen K, Deng K, Wang X, Wang Z, Zheng S, Ren H, He D, Han Y, Asico LD, Jose PA et al. (2015) Activation of D4 dopamine receptor decreases angiotensin II type 1 receptor expression in rat renal proximal tubule cells. Hypertension 65, 153–160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Humphrey JD (2020) Mechanisms of vascular remodeling in hypertension. Am J Hypertens 34, 432–441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Wang L, Zheng J, Du Y, Huang Y, Li J, Liu B, Liu CJ, Zhu Y, Gao Y, Xu Q et al. (2010) Cartilage oligomeric matrix protein maintains the contractile phenotype of vascular smooth muscle cells by interacting with alpha(7)beta(1) integrin. Circ Res 106, 514–525. [DOI] [PubMed] [Google Scholar]
  • 102.Chung CH, Lin KT, Chang CH, Peng HC & Huang TF (2009) The integrin alpha2beta1 agonist, aggretin, promotes proliferation and migration of VSMC through NF-kB translocation and PDGF production. Br J Pharmacol 156, 846–856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Tanigawa K, Maekawa M, Kiyoi T, Nakayama J, Kitazawa R, Kitazawa S, Semba K, Taguchi T, Akita S, Yoshida M et al. (2019) SNX9 determines the surface levels of integrin β1 in vascular endothelial cells: Implication in poor prognosis of human colorectal cancers overexpressing SNX9. J Cell Physiol 234, 17280–17294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Wang HQ, Bai L, Shen BR, Yan ZQ & Jiang ZL (2007) Coculture with endothelial cells enhances vascular smooth muscle cell adhesion and spreading via activation of beta1-integrin and phosphatidylinositol 3-kinase/Akt. Eur J Cell Biol 86, 51–62. [DOI] [PubMed] [Google Scholar]
  • 105.Tseng HY, Thorausch N, Ziegler T, Meves A, Fässler R & Böttcher RT (2014) Sorting nexin 31 binds multiple beta integrin cytoplasmic domains and regulates beta1 integrin surface levels and stability. J Mol Biol 426, 3180–3194. [DOI] [PubMed] [Google Scholar]
  • 106.Mi C, Ye B, Gao Z, Du J, Li R & Huang D (2020) BHLHE40 plays a pathological role in pre-eclampsia through upregulating SNX16 by transcriptional inhibition of miR-196a-5p. Mol Hum Reprod 26, 532–548. [DOI] [PubMed] [Google Scholar]
  • 107.You Y, Zhou C, Li D, Cao ZL, Shen W, Li WZ, Zhang S, Hu B & Shen X (2016) Sorting nexin 10 acting as a novel regulator of macrophage polarization mediates inflammatory response in experimental mouse colitis. Sci Rep 6, 20630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Wen Y & Crowley SD (2020) The varying roles of macrophages in kidney injury and repair. Curr Opin Nephrol Hypertens 29, 286–292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Harwani SC (2018) Macrophages under pressure: the role of macrophage polarization in hypertension. Transl Res 191:45–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.You Y, Bao WL, Zhang SL, Li HD, Li H, Dang WZ, Zou SL, Cao XY, Wang X, Liu LX et al. (2020) Sorting nexin 10 mediates metabolic reprogramming of macrophages in atherosclerosis through the Lyn-dependent TFEB signaling pathway. Circ Res 127, 534–549. [DOI] [PubMed] [Google Scholar]
  • 111.Fan Y, Yang J, Li H, Li H, Zhang S, Li X, Song Y, Dang W, Liu L, Cao X et al. (2020) SNX10 deficiency restricts foam cell formation and protects against atherosclerosis by suppressing CD36-Lyn axis. Can J Cardiol S0828–282X(20)30456–6. doi: 10.1016/j.cjca.2020.05.010. [DOI] [PubMed] [Google Scholar]
  • 112.Williams R, Schlüter T, Roberts MS, Knauth P, Bohnensack R & Cutler DF (2004) Sorting nexin 17 accelerates internalization yet retards degradation of P-selectin. Mol Biol Cell 15, 3095–3105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Knauth P, Schlüter T, Czubayko M, Kirsch C, Florian V, Schreckenberger S, Hahn H & Bohnensack R (2005) Functions of sorting nexin 17 domains and recognition motif for P-selectin trafficking. J Mol Biol 347, 813–825. [DOI] [PubMed] [Google Scholar]
  • 114.Ghai R, Bugarcic A, Liu H, Norwood SJ, Skeldal S, Coulson EJ, Li SS, Teasdale RD & Collins BM (2013) Structural basis for endosomal trafficking of diverse transmembrane cargos by PX-FERM proteins. Proc Natl Acad Sci U S A 110, E643–E652. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Capasso G, Cantone A, Evangelista C, Zacchia M, Trepiccione F, Acone D & Rizzo M (2005) Channels, carriers, and pumps in the pathogenesis of sodium-sensitive hypertension. Semin Nephrol 25, 419–424. [DOI] [PubMed] [Google Scholar]
  • 116.Wichmann L & Althaus M (2020) Evolution of epithelial sodium channels: current concepts and hypotheses. Am J Physiol Regul Integr Comp Physiol. 319, R387–R400. [DOI] [PubMed] [Google Scholar]
  • 117.Ware AW, Rasulov SR, Cheung TT, Lott JS & McDonald FJ (2020) Membrane trafficking pathways regulating the epithelial Na(+) channel. Am J Physiol Renal Physiol 318, F1–F13. [DOI] [PubMed] [Google Scholar]
  • 118.Staruschenko A (2012) Regulation of transport in the connecting tubule and cortical collecting duct. Compr Physiol 2,1541–1584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.McCormick JA & Ellison DH (2015) Distal convoluted tubule. Compr Physiol 5, 45–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Fenton RA & Praetorius J (2011) Molecular physiology of the medullary collecting duct. Compr Physiol 1,1031–1056. [DOI] [PubMed] [Google Scholar]
  • 121.Cheung TT, Geda AC, Ware AW, Rasulov SR, Tenci P, Hamilton KL & McDonald FJ (2020) Retromer is involved in epithelial Na+ channel trafficking. Am J Physiol Renal Physiol 319, F895–F907 [DOI] [PubMed] [Google Scholar]
  • 122.Kato A & Romero MF (2011) Regulation of electroneutral NaCl absorption by the small intestine. Annu Rev Physiol 73, 261–281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Bannert K, Berlin P, Reiner J, Lemcke H, David R, Engelmann R & Lamprecht G (2020) SNX27 regulates DRA activity and mediates its direct recycling by PDZ-interaction in early endosomes at the apical pole of Caco2 cells. Am J Physiol Gastrointest Liver Physiol 318, G854–G869. [DOI] [PubMed] [Google Scholar]
  • 124.Boulkroun S, Ruffieux-Daidié D, Vitagliano JJ, Poirot O, Charles RP, Lagnaz D, Firsov D, Kellenberger S & Staub O (2008) Vasopressin-inducible ubiquitin-specific protease 10 increases ENaC cell surface expression by deubiquitylating and stabilizing sorting nexin 3. Am J Physiol Renal Physiol 295, F889–F900. [DOI] [PubMed] [Google Scholar]
  • 125.Choi HJ, Jang HJ, Park E, Tingskov SJ, Nørregaard R, Jung HJ & Kwon TH (2020) Sorting nexin 27 regulates the lysosomal degradation of aquaporin-2 protein in the kidney collecting duct. Cells 9, 1208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Soleimani M (2015) Insulin resistance and hypertension: new insights. Kidney Int 87, 497–499. [DOI] [PubMed] [Google Scholar]
  • 127.Harashima S, Horiuchi T, Wang Y, Notkins AL, Seino Y & Inagaki N (2012) Sorting nexin 19 regulates the number of dense core vesicles in pancreatic β-cells. J Diabetes Investig 3, 52–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Harashima SI, Harashima C, Nishimura T, Hu Y & Notkins AL (2007) Overexpression of the autoantigen IA-2 puts beta cells into a pre-apoptotic state: autoantigen-induced, but non-autoimmune-mediated, tissue destruction. Clin Exp Immunol 150, 49–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Hu YF, Zhang HL, Cai T, Harashima S & Notkins AL (2005) The IA-2 interactome. Diabetologia 48, 2576–2581. [DOI] [PubMed] [Google Scholar]
  • 130.Buenaventura T, Kanda N, Douzenis PC, Jones B, Bloom SR, Chabosseau P, Corrêa IR Jr, Bosco D, Piemonti L, Marchetti P et al. (2018) A targeted RNAi screen identifies endocytic trafficking factors that control GLP-1 receptor signaling in pancreatic β-Cells. Diabetes 67, 385–399. [DOI] [PubMed] [Google Scholar]
  • 131.Duckworth WC (1988) Insulin degradation: mechanisms, products, and significance. Endocr Rev 9, 319–345. [DOI] [PubMed] [Google Scholar]
  • 132.Valera Mora ME, Scarfone A, Calvani M, Greco AV & Mingrone G (2003) Insulin clearance in obesity. J Am Coll Nutr 22, 487–493. [DOI] [PubMed] [Google Scholar]
  • 133.Tang WJ (2016) Targeting insulin-degrading enzyme to treat type 2 diabetes mellitus. Trends Endocrinol Metab 27, 24–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Li F, Yang J, Villar VAM, Asico LD, Ma X, Armando I, Sanada H, Yoneda M, Felder RA, Jose PA et al. (2018) Loss of renal SNX5 results in impaired IDE activity and insulin resistance in mice. Diabetologia 61, 727–737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Wick KR, Werner ED, Langlais P, Ramos FJ, Dong LQ, Shoelson SE & Liu F (2003) Grb10 inhibits insulin-stimulated insulin receptor substrate (IRS)-phosphatidylinositol 3-kinase/Akt signaling pathway by disrupting the association of IRS-1/IRS-2 with the insulin receptor. J Biol Chem 278, 8460–8467. [DOI] [PubMed] [Google Scholar]
  • 136.Del Aguila LF, Krishnan RK, Ulbrecht JS, Farrell PA, Correll PH, Lang CH, Zierath JR & Kirwan JP (2000) Muscle damage impairs insulin stimulation of IRS-1, PI3-kinase, and Akt-kinase in human skeletal muscle. Am J Physiol Endocrinol Metab 279, E206–E212. [DOI] [PubMed] [Google Scholar]
  • 137.Haft CR, de la Luz Sierra M, Barr VA, Haft DH & Taylor SI (1998) Identification of a family of sorting nexin molecules and characterization of their association with receptors. Mol Cell Biol 18, 7278–7287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Chen Y, Huang L, Qi X & Chen C (2019) Insulin receptor trafficking: consequences for insulin sensitivity and diabetes. Int J Mol Sci 20, 5007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.MaCaulay SL, Stoichevska V, Grusovin J, Gough KH, Castelli LA &Ward CW (2003) Insulin stimulates movement of sorting nexin 9 between cellular compartments: a putative role mediating cell surface receptor expression and insulin action. Biochem J 376, 123–134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Al-Saleem J, Dirksen WP, Martinez MP, Shkriabai N, Kvaratskhelia M, Ratner L, & Green PL (2019) HTLV-1 Tax-1 interacts with SNX27 to regulate cellular localization of the HTLV-1 receptor molecule, GLUT1. PLoS One 14, e0214059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Yang Z, Hong LK, Follett J, Wabitsch M, Hamilton NA, Collins BM, Bugarcic A & Teasdale RD (2016) Functional characterization of retromer in GLUT4 storage vesicle formation and adipocyte differentiation. FASEB J 30, 1037–1050. [DOI] [PubMed] [Google Scholar]
  • 142.Watanabe H, Ogura T, Hosoya M, Kageyama J & Ota Z (1995) Developmental change of kidney dopamine receptors in spontaneously hypertensive rats. Res Commun Mol Pathol Pharmacol 87, 333–344. [PubMed] [Google Scholar]
  • 143.Brismar H, Holtbäck U & Aperia A (2000) Mechanisms by which intrarenal dopamine and ANP interact to regulate sodium metabolism. Clin Exp Hypertens 22, 303–307. [DOI] [PubMed] [Google Scholar]
  • 144.Carey RM, Schoeffel CD, Gildea JJ, Jones JE, McGrath HE, Gordon LN, Park MJ, Sobota RS, Underwood PC, Williams J et al. (2012) Salt sensitivity of blood pressure is associated with polymorphisms in the sodium-bicarbonate cotransporter. Hypertension 60, 1359–1366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Olsson AH, Volkov P, Bacos K, Dayeh T, Hall E, Nilsson EA, Ladenvall C, Rönn T & Ling C (2014) Genome-wide associations between genetic and epigenetic variation influence mRNA expression and insulin secretion in human pancreatic islets. PLoS Genet 10, e1004735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Bao W, Liu X, You Y, Hou H, Wang X, Zhang S, Li H, Feng G, Cao X, Jiang H, et al. (2021) Targeting sorting nexin 10 improves mouse colitis via inhibiting PIKfyve-mediated TBK1/c-Rel signaling activation. Pharmacol Res 169, 105679. [DOI] [PubMed] [Google Scholar]
  • 147.Hu Y, He W, Huang Y, Xiang H, Guo J, Che Y, Cheng X, Hu F, Hu M, Ma T, et al. (2021) Fatty acid synthase-suppressor screening identifies sorting nexin 8 as a therapeutic target for NAFLD. Hepatology doi: 10.1002/hep.32045. Epub ahead of print. [DOI] [PubMed] [Google Scholar]
  • 148.Bao WL, Wu Q, Hu B, Sun D, Zhao S, Shen X, Cheng H & Shen W (2021) Oral nanoparticles of SNX10-shRNA plasmids ameliorate mouse colitis. Int J Nanomedicine. 16, 345–357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Bryant D, Seda M, Peskett E, Maurer C, Pomeranz G, Ghosh M, Hawkins TA, Cleak J, Datta S, Hariri H, et al. (2020) Diverse species-specific phenotypic consequences of loss of function sorting nexin 14 mutations. Sci Rep 10, 13763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Shahin MH, Conrado DJ, Gonzalez D, Gong Y, Lobmeyer MT, Beitelshees AL, Boerwinkle E, Gums JG, Chapman A, Turner ST et al. (2018) Genome-wide association approach identified novel genetic predictors of heart rate response to β-blockers. J Am Heart Assoc 7, e006463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Temkin P, Lauffer B, Jäger S, Cimermancic P, Krogan NJ & von Zastrow M (2011) SNX27 mediates retromer tubule entry and endosome-to-plasma membrane trafficking of signalling receptors. Nat Cell Biol 13, 715–721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Thomopoulos C, Bazoukis G, Tsioufis C & Mancia G (2020) Beta-blockers in hypertension: overview and meta-analysis of randomized outcome trials. J Hypertens 38, 1669–1681. [DOI] [PubMed] [Google Scholar]
  • 153.Li J, Li C, Zhang D, Shi D, Qi M, Feng J, Yuan T, Xu X, Liang D, Xu L, et al. (2014) SNX13 reduction mediates heart failure through degradative sorting of apoptosis repressor with caspase recruitment domain. Nat Commun 5, 5177. [DOI] [PubMed] [Google Scholar]
  • 154.Zhao D, Li X, Liang H, Zheng N, Pan Z, Zhou Y, Liu X, Qian M, Xu B, Zhang Y, et al. (2018) SNX17 produces anti-arrhythmic effects by preserving functional SERCA2a protein in myocardial infarction. Int J Cardiol 272, 298–305. [DOI] [PubMed] [Google Scholar]
  • 155.Lu J, Xu S, Huo Y, Sun D, Hu Y, Wang J, Zhang X, Wang P, Li Z, Liang M, et al. (2021) Sorting nexin 3 induces heart failure via promoting retromer-dependent nuclear trafficking of STAT3. Cell Death Differ 28, 2871–2887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Ish-Shalom E, Meirow Y, Sade-Feldman M, Kanterman J, Wang L, Mizrahi O, Klieger Y & Baniyash M (2016) Impaired SNX9 expression in immune cells during chronic inflammation: prognostic and diagnostic implications. J Immunol 196, 156–167. [DOI] [PubMed] [Google Scholar]
  • 157.Lu X & Crowley SD. (2018) Inflammation in salt-sensitive hypertension and renal damage. Curr Hypertens Rep 20, 103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Jiang X, Konkalmatt P, Yang Y, Gildea J, Jones JE, Cuevas S, Felder RA, Jose PA & Armando I (2014) Single-nucleotide polymorphisms of the dopamine D2 receptor increase inflammation and fibrosis in human renal proximal tubule cells. Hypertension 63, e74–e80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Aguilar-Cazares D, Chavez-Dominguez R, Carlos-Reyes A, Lopez-Camarillo C, Hernadez de la Cruz ON, Lopez-Gonzalez JS (2019) Contribution of angiogenesis to inflammation and cancer. Front Oncol 9, 1399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Qi WX, Fu S, Zhang Q & Guo XM (2016) Incidence and risk of hypertension associated with ramucirumab in cancer patients: a systematic review and meta-analysis. J Cancer Res Ther 12, 775–781. [DOI] [PubMed] [Google Scholar]
  • 161.Essa H, Dobson R, Wright D & Lip GYH (2020) Hypertension management in cardio-oncology. J Hum Hypertens 34, 673–681. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Bian Z, Feng Y, Xue Y, Hu Y, Wang Q, Zhou L, Liu Z, Zhang J, Yin Y, Gu B, et al. (2016) Down-regulation of SNX1 predicts poor prognosis and contributes to drug resistance in colorectal cancer. Tumour Biol 37, 6619–6625. [DOI] [PubMed] [Google Scholar]
  • 163.Zhan XY, Zhang Y, Zhai E, Zhu QY & He Y (2018) Sorting nexin-1 is a candidate tumor suppressor and potential prognostic marker in gastric cancer. Peer J 6, e4829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Nguyen LN, Holdren MS, Nguyen AP, Furuya MH, Bianchini M, Levy E, Mordoh J, Liu A, Guncay GD, Campbell JS, et al. (2006) Sorting nexin 1 down-regulation promotes colon tumorigenesis. Clin Cancer Res 12, 6952–6959. [DOI] [PubMed] [Google Scholar]
  • 165.Zhou Q, Huang T, Jiang Z, Ge C, Chen X, Zhang L, Zhao F, Zhu M, Chen T, Cui Y et al. (2020) Upregulation of SNX5 predicts poor prognosis and promotes hepatocellular carcinoma progression by modulating the EGFR-ERK1/2 signaling pathway. Oncogene 39, 2140–2155 [DOI] [PubMed] [Google Scholar]
  • 166.Uribe ML, Marrocco I & Yarden Y (2021) EGFR in cancer: signaling mechanisms, drugs, and acquired resistance. Cancers (Basel) 13, 2748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Mitsudomi T & Yatabe Y (2010) Epidermal growth factor receptor in relation to tumor development: EGFR gene and cancer. FEBS J 277, 301–308. [DOI] [PubMed] [Google Scholar]
  • 168.Caldieri G, Malabarba MG, Di Fiore PP & Sigismund S (2018) EGFR trafficking in physiology and cancer. Prog Mol Subcell Biol 57, 235–272. [DOI] [PubMed] [Google Scholar]
  • 169.Schwarz DG, Griffin CT, Schneider EA, Yee D & Magnuson T (2002) Genetic analysis of sorting nexins 1 and 2 reveals a redundant and essential function in mice. Mol Biol Cell 13, 3588–3600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Gullapalli A, Garrett TA, Paing MM, Griffin CT, Yang Y & Trejo J (2004) A role for sorting nexin 2 in epidermal growth factor receptor down-regulation: evidence for distinct functions of sorting nexin 1 and 2 in protein trafficking. Mol Biol Cell 15, 2143–2155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Chiow KH, Tan Y, Chua RY, Huang D, Ng ML, Torta F, Wenk MR & Wong SH (2012) SNX3-dependent regulation of epidermal growth factor receptor (EGFR) trafficking and degradation by aspirin in epidermoid carcinoma (A-431) cells. Cell Mol Life Sci 69,1505–1521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Cavet ME, Pang J, Yin G & Berk BC (2008) An epidermal growth factor (EGF) -dependent interaction between GIT1 and sorting nexin 6 promotes degradation of the EGF receptor. FASEB J 22, 3607–3616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Choi JH, Hong WP, Kim MJ, Kim JH, Ryu SH & Suh PG (2004) Sorting nexin 16 regulates EGF receptor trafficking by phosphatidylinositol-3-phosphate interaction with the Phox domain. J Cell Sci 117, 4209–4218. [DOI] [PubMed] [Google Scholar]
  • 174.Nishimura Y, Yoshioka K, Bereczky B & Itoh K (2008) Evidence for efficient phosphorylation of EGFR and rapid endocytosis of phosphorylated EGFR via the early/late endocytic pathway in a gefitinib-sensitive non-small cell lung cancer cell line. Mol Cancer 7, 42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Nishimura Y, Takiguchi S, Yoshioka K, Nakabeppu Y & Itoh K (2012) Silencing of SNX1 by siRNA stimulates the ligand-induced endocytosis of EGFR and increases EGFR phosphorylation in gefitinib-resistant human lung cancer cell lines. Int J Oncol 41,1520–1530. [DOI] [PubMed] [Google Scholar]
  • 176.Nishimura Y, Takiguchi S, Ito S & Itoh K (2015) EGF-stimulated AKT activation is mediated by EGFR recycling via an early endocytic pathway in a gefitinib-resistant human lung cancer cell line. Int J Oncol 46, 1721–1729. [DOI] [PubMed] [Google Scholar]
  • 177.Day TF, Kallakury BVS, Ross JS, Voronel O, Vaidya S, Sheehan CE & Kasid UN (2019) Dual targeting of EGFR and IGF1R in the TNFAIP8 knockdown non-small cell Lung cancer cells. Mol Cancer Res 17, 1207–1219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Ogi S, Fujita H, Kashihara M, Yamamoto C, Sonoda K, Okamoto I, Nakagawa K, Ohdo S, Tanaka Y, Kuwano M et al. (2013) Sorting nexin 2-mediated membrane trafficking of c-Met contributes to sensitivity of molecular-targeted drugs. Cancer Sci 104, 573–583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Lemus-Diaz N, Ferreira RR, Bohnsack KE, Gruber J & Bohnsack MT (2020) The human box C/D snoRNA U3 is a miRNA source and miR-U3 regulates expression of sortin nexin 27. Nucleic Acids Res 48, 8074–8089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Zhang Y, Pang X, Li J, Xu J, Hsu VW & Sun F (2021) Structural insights into membrane remodeling by SNX1. Proc Natl Acad Sci U S A 118, e2022614118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Xu J, Zhang L, Ye Y, Shan Y, Wan C, Wang J, Pei D, Shu X & Liu J (2017) SNX16 regulates the recycling of E-cadherin through a unique mechanism of coordinated membrane and cargo binding. Structure 25,1251–1263.e5. [DOI] [PubMed] [Google Scholar]

RESOURCES