Next Article in Journal
Impact of Phantom Size on Low-Energy Virtual Monoenergetic Images of Three Dual-Energy CT Platforms
Previous Article in Journal
Augmented Reality-Guided Apicoectomy Based on Maxillofacial CBCT Scans
Previous Article in Special Issue
Cardiorenal Syndrome: Ebony and Ivory
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of Semaphorin 3A in Kidney Development and Diseases

1
Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
2
Department of Rheumatology and Immunology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
3
Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
4
Department of Nursing Science, Faculty of Health and Welfare Science, Okayama Prefectural University, Okayama 719-1197, Japan
*
Author to whom correspondence should be addressed.
Diagnostics 2023, 13(19), 3038; https://doi.org/10.3390/diagnostics13193038
Submission received: 23 August 2023 / Revised: 19 September 2023 / Accepted: 21 September 2023 / Published: 25 September 2023

Abstract

:
Kidney diseases are worldwide public health problems affecting millions of people. However, there are still limited therapeutic options against kidney diseases. Semaphorin 3A (SEMA3A) is a secreted and membrane-associated protein, which regulates diverse functions, including immune regulation, cell survival, migration and angiogenesis, thus involving in the several pathogeneses of diseases, including eyes and neurons, as well as kidneys. SEMA3A is expressed in podocytes and tubular cells in the normal adult kidney, and recent evidence has revealed that excess SEMA3A expression and the subsequent signaling pathway aggravate kidney injury in a variety of kidney diseases, including nephrotic syndrome, diabetic nephropathy, acute kidney injury, and chronic kidney disease. In addition, several reports have demonstrated that the inhibition of SEMA3A ameliorated kidney injury via a reduction in cell apoptosis, fibrosis and inflammation; thus, SEMA3A may be a potential therapeutic target for kidney diseases. In this review article, we summarized the current knowledge regarding the role of SEMA3A in kidney pathophysiology and their potential use in kidney diseases.

1. Introduction

Semaphorins are guidance proteins regulating cellular morphology and functions, and, thus, have important roles in developments and diseases, including cancers and metabolic diseases [1]. Semaphorin belongs to the super semaphorin family, consisting of 30 glycoproteins and divided into eight classes [2]. Class 1 and class 2 semaphorins have been identified in invertebrates, and class 3–7 semaphorins in vertebrates. In vertebrates, class 3 semaphorin (SEMA3A-3G) is the only member of secreted proteins [3]. In 1990, Raper et al. isolated a molecule from embryonic chick brain that induced the collapse of neuronal growth cones in culture, and the molecule was originally named Collapsin [4,5], but it was renamed as SEMA3A. Since then, SEMA3A has been well studied not only in axon guidance, but also in pleiotropic functions, including in angiogenesis, immune cell regulation and cell migration [6]. SEMA3A is expressed in kidneys, as well as the nervous system, heart, lung, eyes, bone and immune cells [7,8,9,10,11,12], and regulates tissue development and the maintenance of homeostasis, for example, through the regulation of cell proliferation and migration, and the immune system [13,14,15]. Several reports have identified an increase or decrease in SEMA3A expression under several disease conditions [16,17,18,19]; thus, SEMA3A has been suggested as a possible biomarker, as well as a therapeutic target. For example, it has been reported that retinal SEMA3A expression was increased in a retinal-vein-occlusion mouse model, where the anti-SEMA3A neutralizing antibody BI-X mediated a protective role in intraretinal edema and retinal blood flow [12]. A SEMA3A inhibitor was also reported to protect retinal ganglion cells in animal models of optic nerve injury, retinal ischemia and glaucoma [13]. In a neuron system, the inhibition of SEMA3A enhanced functional recovery during the subacute-stroke recovery period [20]. SEMA3A not only regulated the organization of brain structures affected by autism spectrum disorder (ASD), but also was related to neuron inflammatory processes in ASD [21]. Moreover, lipopolysaccharide (LPS)-induced vascular endothelial cell activation, vascular inflammation, and vascular oxidative stress were substantially improved by the inhibition of SEMA3A using siRNA [22]. The serum SEMA3A level was proposed as a biomarker for diabetic retinopathy in patients with type 2 diabetes [23]. Importantly, it also reflected the severity of diabetic retinopathy, suggesting a value for evaluating patients’ prognosis. A quantitative real-time PCR analysis using glioma tissues verified that SEMA3(A–G) together with six other genes may be useful as biomarkers in the prognosis of a glioma patient’s outcome [24]. Serum SEMA3A was decreased in patients with systemic lupus erythematosus (SLE) and was increased in patients with rheumatoid arthritis and Sjogren’s syndrome, suggesting the important role of SEMA3A in immune-related diseases [25]. SEMA3A expression in cancer tissues was shown to be an independent prognostic factor of overall survival for various types of cancer, such as oral cancer, gastric cancer, breast cancer, prostate cancer, glioblastoma and ovarian cancer. Of note, SEMA3A was significantly correlated with the stage and grade of the disease, depth of invasion, presence of metastases and survival [26,27]. In addition, recent accumulated evidence has also revealed important roles of SEMA3A in kidney diseases.
Kidney diseases, characterized as kidney dysfunction, are a growing public health burden, with a huge economic cost on healthcare systems worldwide [28]. There are several important causes for kidney diseases, including diabetes mellitus (DM), hypertension, glomerulonephritis, obesity and aging. In 2017, a survey showed that 697.5 million cases of all-stage chronic kidney disease (CKD) were recorded, for a global prevalence of 9.1% [29]. Therefore, elucidating the complicated molecular pathology of kidney disease is an urgent task for developing new therapeutic strategies to manage patients with kidney diseases. Over the past few decades, many efforts have been devoted to investigating and evaluating the progression of CKD. One of the important advents for CKD therapy has been the use of sodium–glucose co-transporter-2 (SGLT-2) inhibitor [30,31], which slows down CKD progression. In addition, glucagon-like peptide (GLP) 1 receptor agonists have been reported to reduce the incidences of kidney events in patients with DM [32]. Several promising therapies for CKD, including apoptosis signal-regulating kinase 1 (ASK1) inhibitor, endothelin receptor antagonist, phosphodiesterase inhibitor, Janus kinases (JAK)1/2 inhibitor and nuclear factor erythroid 2-related factor 2 (Nrf2) activator have been undergoing clinical trials [33,34,35,36,37]. Nevertheless, its etiology and pathogenesis are still far from being elucidated. Accumulated evidence has suggested importance roles of SEMA3A signaling in kidney development and several kidney diseases, including proteinuric diseases, acute kidney injury (AKI) and CKD. Several reports have indicated the potential use of inhibitory drugs of SEMA3A signaling against kidney diseases, and also identified SEMA3A expression as a potential biomarker for the early detection and prognosis of AKI and the relapse of nephrotic syndromes. In this review article, we summarize the current body of evidence about SEMA3A in kidney development and diseases.

2. SEMA3A and Its Receptors, and Its Expression in Kidneys

SEMA3A, as with other semaphorin family members, has a conserved N-terminal SEMA domain that contains 500 amino acids, and a diverged C-terminus. The central feature of the SEMA domain is a disulfide-rich seven-blade β-propeller fold. A small cysteine-rich plexin–semaphorin–integrin (PSI) domain is between the SEMA domain and the immunoglobulin (Ig)-like domain. The basic domain is located at the C-terminus of SEMA3A. The C-terminus determines its affinity to neuropilin-1 (NRP1) (Figure 1) [3,38]. SEMA3A has two receptors: NRP1 is necessary for ligand binding, while plexin A is important to subsequent signal transduction. NRP1 is a transmembrane protein with a molecular weight of 120–130 kDa. The extracellular part contains three kinds of domains, made up of two complement binding domains a1/a2, two coagulation factor V/VIII domains, and a c-domain, and is connected to the NRP1 cytoplasmic part (Figure 1) [3]. The signaling receptor Plexin A is a transmembrane glycoprotein with a SEMA3A-like extracellular part, which, implying their common evolutionary origin, is followed by three PSI domains and three IPT (Ig-like, plexins and transcription factors) domains. Importantly, plexin is the only receptor possessing a highly conserved GAP domain (GTPase activating protein), regulating diverse functions in cell activity. Plexin A downstream signaling is complex, involving the following GTPases: R-Ras, M-Ras and Rap (from the Ras family), RhoA, Ras-related C3 botulinum toxin substrate 1 (Rac1), and Rnd (from the Rho family) as well as Fyn phosphorylation [39]. During signal transduction, the SEMA3A–NRP1–Plexin complex forms a holoreceptor [40]. The three-dimensional structure of the SEMA3A–NRP1–Plexin complex has recently been reported [41]. Together, their characteristics add more crosstalk to the pathways involved in SEMA3A signaling. It has been reported that pleiotropic signaling pathways are involved in SEMA3A-–NRP1–Plexin A signaling, including c-Jun N-terminal kinase (JNK) and the Akt signaling pathway.
It has been reported that excess SEMA3A expression may accelerate kidney injury in a variety of kidney diseases via the JNK or Akt signaling pathway [42,43,44,45,46], suggesting that targeting SEMA3A signaling may be a potential therapy against kidney injury. In mammalian adult kidneys, SEMA3A is expressed in podocytes and tubular cells [47], while SEMA3A receptors, NRP1 and Plexin A are expressed throughout life in podocytes, endothelial cells and tubular cells [48,49]. In addition, a recent analysis using a single-cell RNA sequence revealed increased NRP1 expression in activated fibroblasts, suggesting a possible involvement of SEMA3A signaling during the progression of kidney fibrosis [50]. NRP1 is also known as a co-receptor for the vascular endothelial growth factor A isoform 165 (VEGF165); thus, SEMA3A plays a role in suppressing angiogenesis by the competitive inhibition of VEGF signaling through the inhibition of binding to NRP1 [51]. It has been reported that a SEMA3A inhibitor (SM-345431) inhibited connections between SEMA3A and NRP1, thereby blocking SEMA3A signaling [46,52] (Figure 1). Indeed, SM-345431 was shown to antagonize SEMA3A-induced axonal-growth-cone collapse in embryonic neurons [53]. In addition, SM-345431 was shown to preserve the corneal nerve and epithelial integrity in a rodent dry-eye model [54]. To specify an interaction between a peptide inhibitor and the SEMA3A–NRP1 system, a peptide inhibitor was modified with the photoactivatable amino acids-4-benzoyl-l-lphenylalaine or photo-l-leucine using solid-phase peptide synthesis. SEMA3A–peptide interaction was found in a defined area of the SEMA domain, which was also involved in NRP1 [55].

3. SEMA3A in Kidney Development

SEMA3A has roles in axon pathfinding, and cardiovascular, lung and kidney patterning during organogenesis [56,57]. During kidney development, SEMA3A plays an important role in patterning the ureteric bud branching. Recombinant SEMA3A decreases the number of developing glomeruli in vitro and inhibits ureteric bud branching via the downregulation of glial-cell-line-derived neurotrophic factor (GDNF) signaling, competition with VEGF165 and decreased activity of Akt survival pathways. Conversely, the deletion of SEMA3A in mice enhances ureteric bud branching [58]. Thus, SEMA3A functions as a negative regulator of ureteric bud branching during normal kidney development. Reidy et al. established loss- and gain-of-function mouse models, which revealed that SEMA3A−/− mice showed increased endothelial cells and defects in renal vascular patterning, whereas SEMA3A+/+ mice had normal wide-open capillary loops, observed using light microscopy. The morphometric analysis of transmission electron microscopy (TEM) revealed that SEMA3A−/− mice had effaced podocyte foot processes, which were associated with albuminuria. On the other hand, the overexpression of podocyte SEMA3A resulted in glomerular hypoplasia, undifferentiated podocytes and congenital proteinuria [48]. Taken together, a balanced SEMA3A expression may be essential for normal glomerular development, glomerular-filtration-barrier function and ureteric bud branching. In addition to kidney development, the role of SEMA3A in matured kidneys has also been explored. Recombinant SEMA3A injection in adult mice induced nephrotic-range proteinuria [44]. In addition, it was also reported that excess SEMA3A increased starvation-induced apoptosis in cultured podocytes and in the developing kidney in vivo [48,49]. Taken together, SEMA3A not only regulates kidney development, but also may be involved in kidney diseases.
Table 1. Expression of SEMA3A in kidney diseases.
Table 1. Expression of SEMA3A in kidney diseases.
DiseaseEtiologySpeciesSampleSEMA3A
Expression
Ref.
Proteinuric diseasesMCNSHumanUrineIncrease[59]
PANRatsKidneyIncrease[57]
DN-HumanUrineIncrease[60]
db/dbMiceKidneyIncrease[60]
db/dbMiceKidneyIncrease[57]
StreptozotocinMiceKidneyIncrease[61]
AKIIRIMiceKidneyIncrease[62]
LPSMiceKidneyIncrease[45]
ContrastHumanUrineIncrease[63]
Cardiac
operation
HumanSerum/UrineIncrease[43]
CKD-HumanUrineIncrease[64]
LN-HumanKidneyIncrease[65]
-HumanUrineDecrease[66]
DN: diabetic nephropathy; AKI: acute kidney injury; CKD: chronic kidney disease; LN: lupus nephritis; MCNS: minimal-change nephrotic syndrome; PAN: puromycin; IRI: ischemia-reperfusion injury; LPS: lipopolysaccharide.

4. SEMA3A and Kidney Diseases

4.1. Podocytopathy and Diabetic Nephropathy

Podocytes, which have crucial roles in the kidney filtration barrier, line out of the glomerular basement membrane (GBM), prevent urinary protein loss [67]. Podocyte foot processes are linked by slit diaphragms, which regulate cell shape and work as a filtration barrier [68]. Hence, podocyte injury is associated with proteinuria. As indicated above, recombinant SEMA3A injection into adult mice induced nephrotic-range proteinuria [44]; the increase was within 4 h and was resolved within 24 h. TEM analysis revealed the extensive fusion and effacement of podocyte foot processes in kidneys examined 4 h after SEMA3A injection, which were recovered at 48 h, demonstrating that excess circulating SEMA3A may cause podocyte ultrastructural abnormalities, and the permeability of the glomerular filtration barrier is transient and reversible, providing proof of the principle of excess SEMA3A and glomerular disease. In addition, it has also been reported that the SEMA3A-induced downregulation of podocin in a dose-dependent manner decreases the interactions between nephrin, podocin, and CD2-assocaited protein (CD2AP) in cultured podocytes [49]. SEMA3A induced a 10-fold increase in podocyte apoptosis by decreasing the Akt survival pathway [49]. Excess SEMA3A was shown to induce endothelial cell swelling and thickening, lamination of the GBM, and podocyte-foot-process effacement, all of which were transient and reversible upon withdrawal of transgene induction. SEMA3A disrupted podocyte shape in an autocrine fashion, based on podocyte contraction and F-actin collapse [69]. It has been reported that the mechanism under the GBM phenotype change was through increased matrix metalloproteinase 9 expression or the composition of the collagen and laminin chain [47]. Excess SEMA3A caused reversible nephrin downregulation, while the podocin expression and WT1+ nuclei counts were not altered, suggesting that SEMA3A caused decreased nephrin expression without podocyte loss. In addition, GST-binding assays revealed a direct interaction between plexin A and nephrin [69], indicating that extracellular SEMA3A signaling may be directly linked to the slit-diaphragm signaling complex. Increased SEMA3A mRNA and protein expression were found in experimental models of puromycin (PAN)-induced podocyte injury [57] (Table 1). In our previous study, we investigated the pathological roles of SEMA3A signaling on podocyte injury using a doxorubicin (Dox)-induced podocytopathy mouse model and examined the therapeutic effect of a SEMA3A inhibitor, SM-345431 [70] (Table 2). We indicated that Dox induced massive albuminuria and podocyte apoptosis via JNK signaling, as well as an increase in SEMA3A expression in podocytes, all of which were improved by treatments with SM-345431 [46]. We also examined serum and urinary SEMA3A levels in 72 patients who underwent kidney biopsies and showed that urinary SEMA3A levels in minimal-change nephrotic syndrome (MCNS) patients were higher compared to other patients [59]. Furthermore, we evaluated their urinary SEMA3A and MCNS activity, and found that levels of urinary SEMA3A at onset were significantly higher than those at remission in patients with MCNS. These results suggested that the urinary SEMA3A might be useful as a biomarker for MCNS.
Diabetic nephropathy (DN) is one of important complications of DM [71], and is a leading cause of CKD and end-stage renal disease (ESRD) worldwide [72,73]. DN is known to cause podocyte injury and generally starts with microalbuminuria, which progresses to GBM thickness, mesangial expansion, macroalbuminuria, and, finally, a decreasing glomerular filtration rate [74,75]. A multifactorial interaction of factors is involved in DN, such as the formation of advanced glycation end products (AGEs), and the renin–angiotensin system (RAAS), further stimulating protein kinase C, and the generation of reactive oxygen species (ROS) [76,77] and microRNAs. It has been reported that urinary SEMA3A excretion was increased early after the induction of diabetes in diabetic mouse models and in diabetic patients with albuminuria, particularly in those with macroalbuminuria [60]. In diabetic mice, podocyte-specific SEMA3A overexpression (SEMA3A+) caused Kimmelstiel-Wilson-like nodular glomerulosclerosis, massive proteinuria and kidney insufficiency [42]. Increased SEMA3A expression was found in db/db kidneys [57], as well as streptozotocin-induced-diabetes mouse kidneys [61]. The genetic ablation and inhibition of SEMA3A signaling ameliorated diabetes-induced kidney dysfunction [60]. Importantly, a SEMA3A inhibitor, xanthofulvin treatment or the deletion of podocyte plexin A1 abrogated diabetic nodular glomerulosclerosis induced by the SEMA3A+ gain of function [42]. A recent study revealed that microRNAs play important roles in DN pathogenesis. miR-15b-5p restored cell proliferation in high-glucose-induced podocytes by downregulating proapoptotic protein markers, Bax and cleavaged caspase-3, and upregulating the antiapoptotic protein Bcl-2 [78]. miR-15b-5p remarkably decreased the high-glucose-induced inflammatory response via the downregulation of cytokines, IL-1β, TNF-α and IL-6. In addition, it was reported that SEMA3A is a direct target of miR-15b-5p, and the beneficial effects of miR-15b-5p were impeded by excess SEMA3A [78]. SEMA3A is also targeted by miR-23b-3p [79] and miR-16-5p [80]. KCNQ1 opposite-strand/antisense transcript 1(KCNQ1OT1), a long non-coding RNA (lncRNA), was recognized as a miR-23b-3p sponge, and KCNQ1OT1 inhibition ameliorated DN by absorbing miR-23b-3p and regulating SEMA3A [79]. It has also been reported that serum lncRNA T-cell factor7 (TCF7) were elevated in patients with DN and TCF7 silencing ameliorated high-glucose-induced podocyte injury by downregulating SEMA3A via miR-16-5p [80]. Collectively, excess SEMA3A is involved in the progression of DN, and SEMA3A targeting may be a potential therapy against DN.

4.2. Acute Kidney Injury

AKI is recognized as a major public health problem, affecting millions of patients worldwide and leading to higher mortality [81], CKD progression, and sometimes to the new onset of CKD, called the AKI–CKD transition. Kidney ischemia-reperfusion injury (IRI), nephrotoxic agents such as LPS or cisplatin, infection leading to sepsis, and contrast-related injury are major causes of AKI [82]. Currently, AKI is diagnosed according to serum creatinine levels and urine volume [83]. Early prediction before an increase in serum creatinine levels and early treatment are important for patients at risk of AKI. Urinary SEMA3A has been shown to be increased within 6 h after IRI, whereas serum creatinine is increased at 24 h in animals [43]. Urinary SEMA3A was also shown to increase, and peaked at 2 h, after liver-transplantation-induced AKI [84]. Serum SEMA3A in cisplatin-induced AKI was upregulated at 24 h and 48 h. In pediatric patients, AKI was detected 48 h after a cardiopulmonary bypass (CPB) through serum creatinine levels [43], while urine SEMA3A was elevated 2 h after CPB and peaked at 6 h. Moreover, an early increase in urinary SEMA3A levels were associated with clinical outcomes, such as the severity of AKI and the length of a hospital stay [43]. Urinary SEMA3A was compared with other urinary biomarkers, IL-18 [85], L-type fatty acid-binding protein(L-FABP) [86], gelatinase-associated lipocalin (NGAL) [87] and N-acetyl-β-d-glycosaminidase (NAG), in intensive care unit (ICU) admission. These biomarkers showed similar performance in detecting established AKI, later-onset AKI and AKI progression, while urinary SEMA3A was not increased in non-progressive established AKI. Finally, urinary SEMA3A was not increased in sepsis-induced AKI, while levels of other urinary biomarkers were increased [88,89]. An increase in urinary SEMA3A was also reported in contrast-induced acute kidney injury [63]. Among 168 patients who underwent percutaneous coronary intervention (PCI), 20 patients developed AKI. Both urinary SEMA3A and NGAL levels were significantly elevated at 2 h and 6 h post-PCI procedure, and peaked at 2 h post-PCI in the AKI patients, which was much earlier than the rise in serum creatinine levels at 48–72 h post-PCI. Further receiver operating characteristic (ROC) analyses of SEMA3A at 2 h after PCI showed a better predictive sensitivity and specificity compared to NGAL. These results indicate that urinary SEMA3A may be useful as an early and predictive biomarker for AKI.
In addition to biomarkers, SEMA3A signaling is also expected to be a therapeutic target. SEMA3A expression was increased after LPS-induced AKI in mouse tubular epithelial cells, as well as in an LPS-treated rat-kidney-proximal-tubular-epithelial-cell line in vitro via Rac1/nuclear factor kappa-light-chain enhancer of activated B cells (NF-κB) p65 and JNK pathways [45]. In addition, the inhibition of SEMA3A by (-)-epigallocatechin-3-gallate (EGCG) could significantly ameliorate LPS-induced kidney inflammation and apoptosis [45]. It is also reported that genetic silencing and the pharmacological inhibition of SEMA3A ameliorated kidney injury from IRI by inhibiting inflammation and epithelial cell apoptosis [62]. These observations indicate an underling signaling pathway of SEMA3A and the potential utility of a SEMA3A inhibitor as a therapeutic agent for regulating inflammation and apoptosis in AKI. G-protein-coupled receptors (GPCRs) are known to participate in plenty of physiologic functions, and some GPCRs have critical roles in the regulation of kidney function. Among them, Gpr97 is a newly identified adhesion GPCR. Gpr97 was upregulated in IRI-induced AKI mice kidneys [90]. Both in vivo and vitro study have revealed that Gpr97 deficiency attenuated AKI-induced kidney injury by regulating SEMA3A signaling. It was also reported that curcumin, well known for its antioxidant and anti-inflammatory properties, and 12/15 lipoxygenase inhibitor-LOXblock-1 ameliorated IRI-induced AKI by reducing inflammatory processes, oxidative stress and apoptosis, and the effects were through the suppression of the SEMA3A signaling pathway [91]. Another study highlighted the protective effects of human-bone-marrow-derived mesenchymal stem cell exosomes in kidney IRI by delivering miR-199a-3p to kidney cells [92]. The mechanism involved downregulating Sema3A expression and activating Akt and extracellular signal-regulated kinase (ERK) signaling pathways, ultimately leading to reduced apoptosis and improved kidney function. These reports indicate a potential avenue for developing new therapeutic strategies to target SEMA3A signaling for AKI.

4.3. Chronic Kidney Disease

CKD is characterized by progressive damage and a loss of kidney function, in which parenchymal cell loss, chronic inflammation, fibrosis and the reduced regenerative capacity of the kidney are involved in its progression [93]. It was reported that urinary SEMA3A levels were positively correlated with the urine albumin-to-creatinine ratio and serum creatinine levels in hypertensive patients [94]. In the study, patients with CKD showed higher urinary SEMA3A levels compared to those without CKD. Kidney fibrosis is the common pathological pathway of kidney diseases. In our previous study, we evaluated SEMA3A signaling by usingunilateral ureteral obstruction (UUO) mouse model, a kidney fibrosis model [64]. After UUO surgery, SEMA3A expression in the proximal tubular area and NRP1 expression in the fibroblast and tubular cells were increased. The expression of a myofibroblast marker, tenascin-C, and kidney fibrosis were increased in UUO kidneys, all of which were ameliorated by a SEMA3A inhibitor through the regulation of JNK signaling [64]. One of the important mechanisms in kidney tubulointerstitial fibrosis is the kidney tubular epithelial–mesenchymal transition (EMT) process, where kidney tubular epithelial cells lose their cell-to-cell membrane connection and their structural polarity to become a spindle-shaped mesenchymal-like phenotype [95]. Our study indicated that the injection of a SEMA3A inhibitor, SM-345431, could attenuate UUO-induced EMT in vivo [64]. We also demonstrated that recombinant SEMA3A caused tubular cell EMT, and SM-345431 treatment was able to ameliorate TGF-β1-induced EMT in vitro. We also indicated a positive correlation between urinary SEMA3A and a tubular injury marker, urinary NAG, in patients who underwent kidney biopsy. Collectively, SEMA3A signaling may be involved in the progression of kidney fibrosis under CKD, and the inhibition of SEMA3A signaling might be a therapeutic option for protecting a patient from kidney fibrosis [64].

4.4. Systemic Lupus Erythematosus

SEMA3A also regulates immune systems, especially enhancing T-cell and B-cell regulatory properties [3]. Hence, it was reported that SEMA3A is involved in the pathogenesis of autoimmune diseases, including SLE [66], rheumatoid arthritis [96] and Sjogren’s syndrome [97]. SLE is a multi-system autoimmune disease characterized by the aberrant activity of the immune system, and presents with a wide range of clinical manifestations including skin, synovia, brain and kidney [98]. Of note, an analysis of SEMA3A immunostaining in kidneys from patients with lupus nephritis (LN) revealed an increase in SEMA3A expression in patients with LN, while SEMA3A expression was negatively associated with clinical–pathological parameters, including proteinuria and kidney function [65]. In contrast, another study indicated that serum SEMA3A levels in SLE patients were lower than in normal individuals [66]. Aiming to establish a regulatory/protective role for SEMA3A in SLE, serum SEMA3A was assessed in patients with SLE, and this level was compared with SLE disease activity [66], where serum SEMA3A levels were lower in SLE patients compared to those in normal controls. In addition, altered serum SEMA3A levels were found to be inversely correlated with SLE disease activity, mainly with kidney damage and the presence of anti-cardiolipin antibodies. These findings suggest an important role of SEMA3A in SLE.
It was reported that SEMA3A downregulated autoimmune responses by suppressing the over-activity of both B and T cells [99,100]. SEMA3A levels in the B-regulatory cells of patients with SLE were smaller compared to those of normal individuals. Toll-like receptor (TLR)-9 expression could possibly be modulated in the memory B cells of SLE patients, which is associated with the production of inflammatory cytokines such as IL-6 and anti-dsDNA [101,102]. SEMA3A co-cultured with purified B cells from SLE patients significantly reduced TLR-9 expression, supporting the idea that SEMA3A may regulate B-cell autoimmunity in SLE [66]. Serum SEMA3A levels were decreased in SLE while increased in rheumatoid arthritis and Sjogren’s syndrome, compared to healthy controls. How about urinary SEMA3A levels? A study analyzed urinary SEMA3A levels in 38 patients with SLE [103]. Among them, 13 patients had kidney involvement. Urinary SEMA3A levels were lower in SLE patients compared to healthy volunteers, and especially lower in SLE patients with LN than in patients without nephritis, indicating that urinary SEMA3A is inversely correlated with proteinuria and SLE disease activity. The aberrant expression of SEMA3A urine and serum levels in SLE may suggest important roles of SEMA3A in SLE disease activity. Indeed, it was reported that SEMA3A injections in a New Zealand black (NZB)/W mice model of LN delayed the appearance of proteinuria and reduced kidney damage, as well as causing a decrease in immune complex deposition in the glomeruli, indicating the protective effect of SEMA3A in LN [104].
Table 2. Kidney outcome in reports targeting SEMA3A.
Table 2. Kidney outcome in reports targeting SEMA3A.
EtiologyModelSEMA3A-
Targeting
Targeting
Method
SpeciesOutcomeTargetFunctionRef.
Podocyte injuryDoxDownSEMA3A
inhibitor
MiceProteinuria ↓JNKAnti-apoptosis[70]
--UpRecombinant SEMA3AMiceNephrotic
proteinuria
-Podocytopathy[44]
--UpPodocyte
SEMA3A+
MiceProteinuria ↑Dysregulation of nephrin, MMP9, αvβ3 integrinPodocytopathy[69]
DNSTZDownSEMA3A-MiceProteinuria ↓--[60]
STZDownSEMA3A
inhibitor
MiceProteinuria ↓
Kidney fibrosis ↓
Kidney dysfunction ↓
--[60]
High-
glucose
DownmiR-15b-5pPodocyte--Anti-apoptosis
Anti-inflammation
[78]
High-
glucose
UpKCNQ1OT1Podocyte-miR-23b-3pInflammation
Apoptosis
[79]
High-
glucose
DownTCF7 silence/SEMA3A-siRNAPodocyteCytotoxicity↓miR-16-5p-[80]
Podocyte
SEMA3A+
& STZ
DownSEMA3A
inhibitor
(xanthofulvin)
MiceProteinuria ↓
Kidney dysfunction ↓
MICAL1-[42]
AKIIRIDownSEMA3A-/SEMA3A-inhibitorMiceKidney dysfunction ↓
Neutrophil infiltration ↓
-Anti-apoptosis,
Anti-inflammation
[62]
IRI/cisplatinDownGpr97-MiceKidney dysfunction ↓
Neutrophil infiltration ↓
HourAnti-apoptosis,
anti-inflammation
[90]
IRIDownLOXblock-I/CurcuminRats--Anti-apoptosis,
Anti-inflammation
[91]
IRIDownmiR-199a-3pMiceKidney dysfunction ↓Akt/ERK
pathway
Anti-apoptosis[92]
LPSDownEGCGMiceKidney dysfunction ↓
Neutrophil infiltration ↓
Rac1/NF-κB p65/JNK pathwayAnti-apoptosis,
Anti-inflammation
[45]
CKDUUODownSEMA3A
Inhibitor
(SM-345431)
MiceKidney fibrosis ↓JNKAnti-apoptosis,
Anti-EMT
[64]
LNNZBUpRecombinant
SEMA3A
MiceProteinuria ↓
Immune complex-
Deposition ↓
--[104]
DN: diabetic nephropathy; AKI: acute kidney injury; CKD: chronic kidney disease; LN: lupus nephritis; SEMA3A: semaphorin3A; Dox: doxorubicin; STZ: streptozotocin; IRI: ischemia-reperfusion injury; LPS: lipopolysaccharide; UUO: unilateral ureteral obstruction; NZB: New Zealand black mouse; JNK: c-Jun N-terminal kinase; MMP9: matrix metalloproteinase 9; MICAL1: Molecules interaction with CasL1; HuR: RNA-binding protein human antigen R; Akt: activated the protein kinase B; ERK: extracellular signal regulated kinase pathways; Rac1: Ras-related C3 botulinum toxin substrate 1; NF-κB: nuclear factor kappa-light-chain enhancer of activated B cells; EMT: epithelial-mesenchymal transition; KCNQ1OT1: KCNQ1 opposite strand/antisense transcript 1; TCF7: long non-coding RNA T-cell factor 7; EGCG: (-)-epigallocatechin-3-gallate. Up arrows: increase. Down arrows: decrease.

5. SEMA3A and Cardiorenal Syndrome

Cardiorenal syndrome (CRS) is a complex interaction between the heart and kidneys, where dysfunction in one organ causes or exacerbates dysfunction in the other [105]. It was reported that levels of SEMA3A were increased in post-infarcted rat hearts [106]. In the study, intravenous SEMA3A administration improved cardiac autonomic regulation, while the infarct size and cardiac function were not affected. It was also reported that SEMA3A expression in circulating monocytes was increased in patients 30 days after myocardial infarction [107]. In the report, SEMA3A reduced cardiac inflammation after myocardial ischemia in mice. Although further studies are still required to conclude, SEMA3A signaling might be involved in the CRS after myocardial infarction.

6. Conclusions

We have summarized the current knowledge regarding the role of SEMA3A in kidney pathophysiology and its potential use in kidney diseases. On the basis of these studies, SEMA3A plays an important role in kidney morphogenesis and kidney diseases. SEMA3A loss-of-function studies indicate that SEMA3A is required for the maintenance of structure and function in the glomerular filtration barrier. In contrast, excess SEMA3A causes the progression of a variety of kidney diseases, including DN, AKI and CKD, through an increase in albuminuria, kidney fibrosis, apoptosis and inflammation. Current speculations around SEMA3A and the pathophysiology of kidney diseases is summarized in Figure 2. Increased SEMA3A expression from tubular cells may cause tubular apoptosis and EMT in an autocrine manner under AKI and CKD, as well as cause fibroblast activation. In addition, increased SEMA3A expression from podocytes may cause podocytopathy, leading to proteinuria in DN. Therefore, SEMA3A-targeting therapy may be a novel therapeutic option for treatment against a variety of kidney diseases, including AKI, CKD and proteinuric diseases. Indeed, SEMA3A-mutant mice or the pharmacological-based inhibition of SEMA3A protected from these kidney diseases, suggesting the potential of SEMA3A inhibitory treatment in clinic use in the future. On the other hand, SEMA3A deficiency may lead to the progression of LN through upregulating autoimmune responses by the activation of T cells and B cells, suggesting a concern of side effects of the inflammatory response in kidneys under SEMA3A inhibitory therapy. In addition, the detailed mechanisms of SEMA3A underlying kidney diseases are not fully understood;, there are several possible signaling pathways, involving SEMA3A signaling, such as Rac1/NF-κB p65, JNK and TLR4 signaling, which may regulate inflammation and cell apoptosis. Of note, the Rho family of GTPases acts downstream of plexin A, regulating adhesion, proliferation, migration and survival in different cell types, and interacts with diverse signaling pathways, suggesting that the Rho family may be involved as a downstream effector of SEMA3A. We previously proposed a possible connection between the SEMA3A–NRP1–plexin A1 complex and JNK signaling with FERM, ARH/RhoGEF and the pleckstrin domain protein 2 (FARP2)/Rac1/Mixed-LineageKinase3 (MLK3)/mitogen-activated protein kinase (MKK) 4/7 cascade [64]. Further studies are still needed for a deep understanding of SEMA3A signaling.

Author Contributions

Conceptualization, K.T.; formal analysis, Y.S., K.T., H.N. and K.F.; investigation, K.T., H.N. and K.F.; writing—original draft preparation, Y.S. and K.T.; writing—review and editing, K.T., H.N., K.F., S.K. and J.W.; supervision, J.W. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Japanese Society for the Promotion of Science (JSPS)/Grants-in-Aid for Young Scientists 18K15978 and 20K17283 (to K.T.) and Shanghai Pujiang Young Rheumatologists Training Program (SPROG2201, to Y.S.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

There were no datasets generated during and/or analyzed during the current study.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Lu, Q.; Zhu, L. The Role of Semaphorins in Metabolic Disorders. Int. J. Mol. Sci. 2020, 21, 5641. [Google Scholar] [CrossRef]
  2. Alto, L.T.; Terman, J.R. Semaphorins and their Signaling Mechanisms. Methods Mol. Biol. 2017, 1493, 1–25. [Google Scholar]
  3. Kiseleva, E.P.; Rutto, K.V. Semaphorin 3A in the Immune System: Twenty Years of Study. Biochemistry 2022, 87, 640–657. [Google Scholar] [CrossRef] [PubMed]
  4. Raper, J.A.; Kapfhammer, J.P. The enrichment of a neuronal growth cone collapsing activity from embryonic chick brain. Neuron 1990, 4, 21–29. [Google Scholar] [CrossRef] [PubMed]
  5. Luo, Y.; Raible, D.; Raper, J.A. Collapsin: A protein in brain that induces the collapse and paralysis of neuronal growth cones. Cell 1993, 75, 217–227. [Google Scholar] [CrossRef]
  6. Roth, L.; Koncina, E.; Satkauskas, S.; Cremel, G.; Aunis, D.; Bagnard, D. The many faces of semaphorins: From development to pathology. Cell Mol. Life Sci. 2009, 66, 649–666. [Google Scholar] [CrossRef]
  7. Kim, O.; Tran, P.T.; Gal, M.; Lee, S.J.; Na, S.H.; Hwangbo, C.; Lee, J.H. RAS-stimulated release of exosomal miR-494-3p promotes the osteolytic bone metastasis of breast cancer cells. Int. J. Mol. Med. 2023, 52, 84. [Google Scholar] [CrossRef] [PubMed]
  8. Kurokawa, S.; Kashimoto, M.; Hagikura, K.; Shimodai-Yamada, S.; Otsuka, N.; Wakamatsu, Y.; Nagashima, K.; Matsumoto, T.; Hao, H.; Okumura, Y. Intravenous Semaphorin 3A Administration Maintains Cardiac Contractility and Improves Electrical Remodeling in a Mouse Model of Isoproterenol-Induced Heart Failure. Int. Heart J. 2023, 64, 453–461. [Google Scholar] [CrossRef]
  9. Qiu, Q.; Yu, X.; Chen, Q.; He, X. Sema3A inactivates the ERK/JNK signalling pathways to alleviate inflammation and oxidative stress in lipopolysaccharide-stimulated rat endothelial cells and lung tissues. Autoimmunity 2023, 56, 2200908. [Google Scholar] [CrossRef]
  10. Tian, T.; Chen, L.; Wang, Z.; Zhu, M.; Xu, W.; Wu, B. Sema3A Drives Alternative Macrophage Activation in the Resolution of Periodontitis via PI3K/AKT/mTOR Signaling. Inflammation 2023, 46, 876–891. [Google Scholar] [CrossRef]
  11. Ieguchi, K.; Funakoshi, M.; Mishima, T.; Takizawa, K.; Omori, T.; Nakamura, F.; Watanabe, M.; Tsuji, M.; Kiuchi, Y.; Kobayashi, S.; et al. The Sympathetic Nervous System Contributes to the Establishment of Pre-Metastatic Pulmonary Microenvironments. Int. J. Mol. Sci. 2022, 23, 10652. [Google Scholar] [CrossRef] [PubMed]
  12. Nakamura, S.; Nishinaka, A.; Hidaka, Y.; Shimazawa, M.; Thomas, L.; Bakker, R.A.; Hara, H. Efficacy of an Anti-Semaphorin 3A Neutralizing Antibody in a Male Experimental Retinal Vein Occlusion Mouse Model. Investig. Ophthalmol. Vis. Sci. 2022, 63, 14. [Google Scholar] [CrossRef] [PubMed]
  13. Nitzan, A.; Corredor-Sanchez, M.; Galron, R.; Nahary, L.; Safrin, M.; Bruzel, M.; Moure, A.; Bonet, R.; Perez, Y.; Bujons, J.; et al. Inhibition of Sema-3A Promotes Cell Migration, Axonal Growth, and Retinal Ganglion Cell Survival. Transl. Vis. Sci. Technol. 2021, 10, 16. [Google Scholar] [CrossRef] [PubMed]
  14. Kanth, S.M.; Gairhe, S.; Torabi-Parizi, P. The Role of Semaphorins and Their Receptors in Innate Immune Responses and Clinical Diseases of Acute Inflammation. Front. Immunol. 2021, 12, 672441. [Google Scholar] [CrossRef] [PubMed]
  15. Higgins, D.M.O.; Caliva, M.; Schroeder, M.; Carlson, B.; Upadhyayula, P.S.; Milligan, B.D.; Cheshier, S.H.; Weissman, I.L.; Sarkaria, J.N.; Meyer, F.B.; et al. Semaphorin 3A mediated brain tumor stem cell proliferation and invasion in EGFRviii mutant gliomas. BMC Cancer 2020, 20, 1213. [Google Scholar] [CrossRef] [PubMed]
  16. Karpuz, T.; Araz, M.; Korkmaz, L.; Kilinc, I.; Findik, S.; Karaagac, M.; Eryilmaz, M.K.; Artac, M. The Prognostic Value of Serum Semaphorin3A and VEGF Levels in Patients with Metastatic Colorectal Cancer. J. Gastrointest. Cancer 2020, 51, 491–497. [Google Scholar] [CrossRef]
  17. Sabag, A.D.; Dias-Polak, D.; Bejar, J.; Sheffer, H.; Bergman, R.; Vadasz, Z. Altered expression of regulatory molecules in the skin of psoriasis. Immunol. Res. 2018, 66, 649–654. [Google Scholar] [CrossRef]
  18. Gao, H.; Ma, X.X.; Guo, Q.; Xie, L.F.; Zhong, Y.C.; Zhang, X.W. Expression of circulating Semaphorin3A and its association with inflammation and bone destruction in rheumatoid arthritis. Clin. Rheumatol. 2018, 37, 2073–2080. [Google Scholar] [CrossRef]
  19. Jacob, S.; Al-Kandari, A.; Alroughani, R.; Al-Temaimi, R. Assessment of plasma biomarkers for their association with Multiple Sclerosis progression. J. Neuroimmunol. 2017, 305, 5–8. [Google Scholar] [CrossRef]
  20. Hira, K.; Ueno, Y.; Tanaka, R.; Miyamoto, N.; Yamashiro, K.; Inaba, T.; Urabe, T.; Okano, H.; Hattori, N. Astrocyte-Derived Exosomes Treated with a Semaphorin 3A Inhibitor Enhance Stroke Recovery via Prostaglandin D2 Synthase. Stroke 2018, 49, 2483–2494. [Google Scholar] [CrossRef]
  21. Matrone, C.; Ferretti, G. Semaphorin 3A influences neuronal processes that are altered in patients with autism spectrum disorder: Potential diagnostic and therapeutic implications. Neurosci. Biobehav. Rev. 2023, 153, 105338. [Google Scholar] [CrossRef]
  22. Fu, Y.; Liu, J.W.; Wu, J.; Wu, Z.X.; Li, J.; Ji, H.F.; Liang, N.P.; Zhang, H.J.; Lai, Z.Q.; Dong, Y.F. Inhibition of semaphorin-3a alleviates lipopolysaccharide-induced vascular injury. Microvasc. Res. 2022, 142, 104346. [Google Scholar] [CrossRef] [PubMed]
  23. Mokhtar, E.R.; Mahmoud, D.A.; Ebrahim, G.E.; Al Anany, M.G.; Seliem, N.; Hassan, M.M. Serum metabolomic profiles and semaphorin-3A as biomarkers of diabetic retinopathy progression. Egypt. J. Immunol. 2023, 30, 83–98. [Google Scholar] [CrossRef]
  24. Valiulyte, I.; Steponaitis, G.; Kardonaite, D.; Tamasauskas, A.; Kazlauskas, A. A SEMA3 Signaling Pathway-Based Multi-Biomarker for Prediction of Glioma Patient Survival. Int. J. Mol. Sci. 2020, 21, 7396. [Google Scholar] [CrossRef] [PubMed]
  25. Wang, P.; Mao, Y.M.; Liu, L.N.; Zhao, C.N.; Li, X.M.; Pan, H.F. Decreased Expression of Semaphorin 3A and Semaphorin 7A Levels and Its Association with Systemic Lupus Erythematosus. Immunol. Investig. 2020, 49, 69–80. [Google Scholar] [CrossRef] [PubMed]
  26. Izycka, N.; Sterzynska, K.; Januchowski, R.; Nowak-Markwitz, E. Semaphorin 3A (SEMA3A), protocadherin 9 (PCdh9), and S100 calcium binding protein A3 (S100A3) as potential biomarkers of carcinogenesis and chemoresistance of different neoplasms, including ovarian cancer—Review of literature. Ginekol. Pol. 2019, 90, 223–227. [Google Scholar] [CrossRef]
  27. Jiang, H.; Qi, L.; Wang, F.; Sun, Z.; Huang, Z.; Xi, Q. Decreased semaphorin 3A expression is associated with a poor prognosis in patients with epithelial ovarian carcinoma. Int. J. Mol. Med. 2015, 35, 1374–1380. [Google Scholar] [CrossRef]
  28. Luyckx, V.A.; Tonelli, M.; Stanifer, J.W. The global burden of kidney disease and the sustainable development goals. Bull. World Health Organ. 2018, 96, 414–422D. [Google Scholar] [CrossRef]
  29. GBD Chronic Kidney Disease Collaboration. Global, regional, and national burden of chronic kidney disease, 1990–2017: A systematic analysis for the Global Burden of Disease Study 2017. Lancet 2020, 395, 709–733. [Google Scholar] [CrossRef]
  30. EMPA-Kidney Collaborative Group; Herrington, W.G.; Staplin, N.; Wanner, C.; Green, J.B.; Hauske, S.J.; Emberson, J.R.; Preiss, D.; Judge, P.; Mayne, K.J.; et al. Empagliflozin in Patients with Chronic Kidney Disease. N. Engl. J. Med. 2023, 388, 117–127. [Google Scholar]
  31. Heerspink, H.J.L.; Stefansson, B.V.; Correa-Rotter, R.; Chertow, G.M.; Greene, T.; Hou, F.F.; Mann, J.F.E.; McMurray, J.J.V.; Lindberg, M.; Rossing, P.; et al. Dapagliflozin in Patients with Chronic Kidney Disease. N. Engl. J. Med. 2020, 383, 1436–1446. [Google Scholar] [CrossRef]
  32. Prattichizzo, F.; de Candia, P.; Ceriello, A. Diabetes and kidney disease: Emphasis on treatment with SGLT-2 inhibitors and GLP-1 receptor agonists. Metabolism 2021, 120, 154799. [Google Scholar] [CrossRef]
  33. Nangaku, M.; Takama, H.; Ichikawa, T.; Mukai, K.; Kojima, M.; Suzuki, Y.; Watada, H.; Wada, T.; Ueki, K.; Narita, I.; et al. Randomized, double-blind, placebo-controlled phase 3 study of bardoxolone methyl in patients with diabetic kidney disease: Design and baseline characteristics of the AYAME study. Nephrol. Dial. Transplant. 2023, 38, 1204–1216. [Google Scholar] [CrossRef]
  34. Leehey, D.J.; Carlson, K.; Reda, D.J.; Craig, I.; Clise, C.; Conner, T.A.; Agarwal, R.; Kaufman, J.S.; Anderson, R.J.; Lammie, D.; et al. Pentoxifylline in diabetic kidney disease (VA PTXRx): Protocol for a pragmatic randomised controlled trial. BMJ Open 2021, 11, e053019. [Google Scholar] [CrossRef]
  35. Chertow, G.M.; Pergola, P.E.; Chen, F.; Kirby, B.J.; Sundy, J.S.; Patel, U.D.; GS-US-223-1015 Investigators. Effects of Selonsertib in Patients with Diabetic Kidney Disease. J. Am. Soc. Nephrol. 2019, 30, 1980–1990. [Google Scholar] [CrossRef]
  36. Tuttle, K.R.; Brosius, F.C., 3rd; Adler, S.G.; Kretzler, M.; Mehta, R.L.; Tumlin, J.A.; Tanaka, Y.; Haneda, M.; Liu, J.; Silk, M.E.; et al. JAK1/JAK2 inhibition by baricitinib in diabetic kidney disease: Results from a Phase 2 randomized controlled clinical trial. Nephrol. Dial. Transplant. 2018, 33, 1950–1959. [Google Scholar] [CrossRef]
  37. Lin, C.W.; Mostafa, N.M.; Andress, D.L.; Brennan, J.J.; Klein, C.E.; Awni, W.M. Relationship Between Atrasentan Concentrations and Urinary Albumin to Creatinine Ratio in Western and Japanese Patients with Diabetic Nephropathy. Clin. Ther. 2018, 40, 242–251. [Google Scholar] [CrossRef]
  38. Toledano, S.; Nir-Zvi, I.; Engelman, R.; Kessler, O.; Neufeld, G. Class-3 Semaphorins and Their Receptors: Potent Multifunctional Modulators of Tumor Progression. Int. J. Mol. Sci. 2019, 20, 556. [Google Scholar] [CrossRef]
  39. Tamagnone, L.; Artigiani, S.; Chen, H.; He, Z.; Ming, G.I.; Song, H.; Chedotal, A.; Winberg, M.L.; Goodman, C.S.; Poo, M.; et al. Plexins are a large family of receptors for transmembrane, secreted, and GPI-anchored semaphorins in vertebrates. Cell 1999, 99, 71–80. [Google Scholar] [CrossRef]
  40. Janssen, B.J.; Malinauskas, T.; Weir, G.A.; Cader, M.Z.; Siebold, C.; Jones, E.Y. Neuropilins lock secreted semaphorins onto plexins in a ternary signaling complex. Nat. Struct. Mol. Biol. 2012, 19, 1293–1299. [Google Scholar] [CrossRef]
  41. Lu, D.; Shang, G.; He, X.; Bai, X.C.; Zhang, X. Architecture of the Sema3A/PlexinA4/Neuropilin tripartite complex. Nat. Commun. 2021, 12, 3172. [Google Scholar] [CrossRef]
  42. Aggarwal, P.K.; Veron, D.; Thomas, D.B.; Siegel, D.; Moeckel, G.; Kashgarian, M.; Tufro, A. Semaphorin3a promotes advanced diabetic nephropathy. Diabetes 2015, 64, 1743–1759. [Google Scholar] [CrossRef]
  43. Jayakumar, C.; Ranganathan, P.; Devarajan, P.; Krawczeski, C.D.; Looney, S.; Ramesh, G. Semaphorin 3A is a new early diagnostic biomarker of experimental and pediatric acute kidney injury. PLoS ONE 2013, 8, e58446. [Google Scholar] [CrossRef]
  44. Tapia, R.; Guan, F.; Gershin, I.; Teichman, J.; Villegas, G.; Tufro, A. Semaphorin3a disrupts podocyte foot processes causing acute proteinuria. Kidney Int. 2008, 73, 733–740. [Google Scholar] [CrossRef]
  45. Tian, X.; Gan, H.; Zeng, Y.; Zhao, H.; Tang, R.; Xia, Y. Inhibition of semaphorin-3a suppresses lipopolysaccharide-induced acute kidney injury. J. Mol. Med. 2018, 96, 713–724. [Google Scholar] [CrossRef]
  46. Sang, Y.; Tsuji, K.; Inoue-Torii, A.; Fukushima, K.; Kitamura, S.; Wada, J. Semaphorin3A-Inhibitor Ameliorates Doxorubicin-Induced Podocyte Injury. Int. J. Mol. Sci. 2020, 21, 4099. [Google Scholar] [CrossRef]
  47. Tufro, A. Semaphorin3a signaling, podocyte shape, and glomerular disease. Pediatr. Nephrol. 2014, 29, 751–755. [Google Scholar] [CrossRef]
  48. Reidy, K.J.; Villegas, G.; Teichman, J.; Veron, D.; Shen, W.; Jimenez, J.; Thomas, D.; Tufro, A. Semaphorin3a regulates endothelial cell number and podocyte differentiation during glomerular development. Development 2009, 136, 3979–3989. [Google Scholar] [CrossRef]
  49. Guan, F.; Villegas, G.; Teichman, J.; Mundel, P.; Tufro, A. Autocrine class 3 semaphorin system regulates slit diaphragm proteins and podocyte survival. Kidney Int. 2006, 69, 1564–1569. [Google Scholar] [CrossRef]
  50. Wu, H.; Kirita, Y.; Donnelly, E.L.; Humphreys, B.D. Advantages of Single-Nucleus over Single-Cell RNA Sequencing of Adult Kidney: Rare Cell Types and Novel Cell States Revealed in Fibrosis. J. Am. Soc. Nephrol. 2019, 30, 23–32. [Google Scholar] [CrossRef]
  51. Kolodkin, A.L.; Levengood, D.V.; Rowe, E.G.; Tai, Y.T.; Giger, R.J.; Ginty, D.D. Neuropilin is a semaphorin III receptor. Cell 1997, 90, 753–762. [Google Scholar] [CrossRef]
  52. Omoto, M.; Yoshida, S.; Miyashita, H.; Kawakita, T.; Yoshida, K.; Kishino, A.; Kimura, T.; Shibata, S.; Tsubota, K.; Okano, H.; et al. The semaphorin 3A inhibitor SM-345431 accelerates peripheral nerve regeneration and sensitivity in a murine corneal transplantation model. PLoS ONE 2012, 7, e47716. [Google Scholar] [CrossRef]
  53. Ivakhnitskaia, E.; Chin, M.R.; Siegel, D.; Guaiquil, V.H. Vinaxanthone inhibits Semaphorin3A induced axonal growth cone collapse in embryonic neurons but fails to block its growth promoting effects on adult neurons. Sci. Rep. 2021, 11, 13019. [Google Scholar] [CrossRef]
  54. Yamazaki, R.; Yamazoe, K.; Yoshida, S.; Hatou, S.; Inagaki, E.; Okano, H.; Tsubota, K.; Shimmura, S. The Semaphorin 3A inhibitor SM-345431 preserves corneal nerve and epithelial integrity in a murine dry eye model. Sci. Rep. 2017, 7, 15584. [Google Scholar] [CrossRef]
  55. Kretschmer, K.; Stichel, J.; Bellmann-Sickert, K.; Baumann, L.; Bierer, D.; Riedl, B.; Beck-Sickinger, A.G. Pinpointing the interaction site between semaphorin-3A and its inhibitory peptide. J. Pept. Sci. 2023, 29, e3460. [Google Scholar] [CrossRef]
  56. Tran, T.S.; Kolodkin, A.L.; Bharadwaj, R. Semaphorin regulation of cellular morphology. Annu. Rev. Cell Dev. Biol. 2007, 23, 263–292. [Google Scholar] [CrossRef]
  57. Reidy, K.; Tufro, A. Semaphorins in kidney development and disease: Modulators of ureteric bud branching, vascular morphogenesis, and podocyte-endothelial crosstalk. Pediatr. Nephrol. 2011, 26, 1407–1412. [Google Scholar] [CrossRef]
  58. Tufro, A.; Teichman, J.; Woda, C.; Villegas, G. Semaphorin3a inhibits ureteric bud branching morphogenesis. Mech. Dev. 2008, 125, 558–568. [Google Scholar] [CrossRef]
  59. Inoue-Torii, A.; Kitamura, S.; Wada, J.; Tsuji, K.; Makino, H. The level of urinary semaphorin3A is associated with disease activity in patients with minimal change nephrotic syndrome. Int. J. Nephrol. Renovasc. Dis. 2017, 10, 167–174. [Google Scholar] [CrossRef]
  60. Mohamed, R.; Ranganathan, P.; Jayakumar, C.; Nauta, F.L.; Gansevoort, R.T.; Weintraub, N.L.; Brands, M.; Ramesh, G. Urinary semaphorin 3A correlates with diabetic proteinuria and mediates diabetic nephropathy and associated inflammation in mice. J. Mol. Med. 2014, 92, 1245–1256. [Google Scholar] [CrossRef]
  61. Veron, D.; Bertuccio, C.A.; Marlier, A.; Reidy, K.; Garcia, A.M.; Jimenez, J.; Velazquez, H.; Kashgarian, M.; Moeckel, G.W.; Tufro, A. Podocyte vascular endothelial growth factor (Vegf164) overexpression causes severe nodular glomerulosclerosis in a mouse model of type 1 diabetes. Diabetologia 2011, 54, 1227–1241. [Google Scholar] [CrossRef]
  62. Ranganathan, P.; Jayakumar, C.; Mohamed, R.; Weintraub, N.L.; Ramesh, G. Semaphorin 3A inactivation suppresses ischemia-reperfusion-induced inflammation and acute kidney injury. Am. J. Physiol. Ren. Physiol. 2014, 307, F183–F194. [Google Scholar] [CrossRef]
  63. Ning, L.; Li, Z.; Wei, D.; Chen, H.; Yang, C.; Wu, D.; Wang, Y.; Zhang, J. Urinary semaphorin 3A as an early biomarker to predict contrast-induced acute kidney injury in patients undergoing percutaneous coronary intervention. Braz. J. Med. Biol. Res. 2018, 51, e6487. [Google Scholar] [CrossRef]
  64. Sang, Y.; Tsuji, K.; Fukushima, K.; Takahashi, K.; Kitamura, S.; Wada, J. Semaporin3A inhibitor ameliorates renal fibrosis through the regulation of JNK signaling. Am. J. Physiol. Ren. Physiol. 2021, 321, F740–F756. [Google Scholar] [CrossRef]
  65. Vadasz, Z.; Ben-Izhak, O.; Bejar, J.; Sabo, E.; Kessel, A.; Storch, S.; Toubi, E. The involvement of immune semaphorins and neuropilin-1 in lupus nephritis. Lupus 2011, 20, 1466–1473. [Google Scholar] [CrossRef]
  66. Vadasz, Z.; Toubi, E. Semaphorin 3A—A marker for disease activity and a potential putative disease-modifying treatment in systemic lupus erythematosus. Lupus 2012, 21, 1266–1270. [Google Scholar] [CrossRef]
  67. Koehler, S.; Miner, J.H.; Staruschenko, A. Call for papers: Podocyte physiology and pathophysiology. Am. J. Physiol. Ren. Physiol. 2023, 324, F505–F510. [Google Scholar] [CrossRef]
  68. Tufro, A. Podocyte Shape Regulation by Semaphorin 3A and MICAL-1. Methods Mol. Biol. 2017, 1493, 393–399. [Google Scholar]
  69. Reidy, K.J.; Aggarwal, P.K.; Jimenez, J.J.; Thomas, D.B.; Veron, D.; Tufro, A. Excess podocyte semaphorin-3A leads to glomerular disease involving plexinA1-nephrin interaction. Am. J. Pathol. 2013, 183, 1156–1168. [Google Scholar] [CrossRef]
  70. Kumagai, K.; Hosotani, N.; Kikuchi, K.; Kimura, T.; Saji, I. Xanthofulvin, a novel semaphorin inhibitor produced by a strain of Penicillium. J. Antibiot. 2003, 56, 610–616. [Google Scholar] [CrossRef]
  71. Samsu, N. Diabetic Nephropathy: Challenges in Pathogenesis, Diagnosis, and Treatment. BioMed Res. Int. 2021, 2021, 1497449. [Google Scholar] [CrossRef]
  72. Valencia, W.M.; Florez, H. How to prevent the microvascular complications of type 2 diabetes beyond glucose control. BMJ 2017, 356, i6505. [Google Scholar] [CrossRef]
  73. Burrows, N.R.; Hora, I.; Geiss, L.S.; Gregg, E.W.; Albright, A. Incidence of End-Stage Renal Disease Attributed to Diabetes Among Persons with Diagnosed Diabetes—United States and Puerto Rico, 2000–2014. Morb. Mortal. Wkly. Rep. 2017, 66, 1165–1170. [Google Scholar] [CrossRef]
  74. Lane, P.H.; Steffes, M.W.; Mauer, S.M. Renal histologic changes in diabetes mellitus. Semin. Nephrol. 1990, 10, 254–259. [Google Scholar]
  75. Maezawa, Y.; Takemoto, M.; Yokote, K. Cell biology of diabetic nephropathy: Roles of endothelial cells, tubulointerstitial cells and podocytes. J. Diabetes Investig. 2015, 6, 3–15. [Google Scholar] [CrossRef]
  76. Cao, Z.; Cooper, M.E. Pathogenesis of diabetic nephropathy. J. Diabetes Investig. 2011, 2, 243–247. [Google Scholar] [CrossRef]
  77. Kumar Pasupulati, A.; Chitra, P.S.; Reddy, G.B. Advanced glycation end products mediated cellular and molecular events in the pathology of diabetic nephropathy. Biomol. Concepts 2016, 7, 293–309. [Google Scholar] [CrossRef]
  78. Fu, Y.; Wang, C.; Zhang, D.; Chu, X.; Zhang, Y.; Li, J. miR-15b-5p ameliorated high glucose-induced podocyte injury through repressing apoptosis, oxidative stress, and inflammatory responses by targeting Sema3A. J. Cell. Physiol. 2019, 234, 20869–20878. [Google Scholar] [CrossRef]
  79. Fei, B.; Zhou, H.; He, Z.; Wang, S. KCNQ1OT1 inhibition alleviates high glucose-induced podocyte injury by adsorbing miR-23b-3p and regulating Sema3A. Clin. Exp. Nephrol. 2022, 26, 385–397. [Google Scholar] [CrossRef]
  80. Jiang, Z.; Qian, L.; Yang, R.; Wu, Y.; Guo, Y.; Chen, T. LncRNA TCF7 contributes to high glucose-induced damage in human podocytes by up-regulating SEMA3A via sponging miR-16-5p. J. Diabetes Investig. 2023, 14, 193–204. [Google Scholar] [CrossRef]
  81. Singbartl, K.; Kellum, J.A. AKI in the ICU: Definition, epidemiology, risk stratification, and outcomes. Kidney Int. 2012, 81, 819–825. [Google Scholar] [CrossRef] [PubMed]
  82. Jang, H.R.; Rabb, H. Immune cells in experimental acute kidney injury. Nat. Rev. Nephrol. 2015, 11, 88–101. [Google Scholar] [CrossRef] [PubMed]
  83. Kellum, J.A.; Romagnani, P.; Ashuntantang, G.; Ronco, C.; Zarbock, A.; Anders, H.J. Acute kidney injury. Nat. Rev. Dis. Primers 2021, 7, 52. [Google Scholar] [CrossRef]
  84. Lewandowska, L.; Matuszkiewicz-Rowinska, J.; Jayakumar, C.; Oldakowska-Jedynak, U.; Looney, S.; Galas, M.; Dutkiewicz, M.; Krawczyk, M.; Ramesh, G. Netrin-1 and semaphorin 3A predict the development of acute kidney injury in liver transplant patients. PLoS ONE 2014, 9, e107898. [Google Scholar] [CrossRef] [PubMed]
  85. Parikh, C.R.; Mishra, J.; Thiessen-Philbrook, H.; Dursun, B.; Ma, Q.; Kelly, C.; Dent, C.; Devarajan, P.; Edelstein, C.L. Urinary IL-18 is an early predictive biomarker of acute kidney injury after cardiac surgery. Kidney Int. 2006, 70, 199–203. [Google Scholar] [CrossRef] [PubMed]
  86. Doi, K.; Negishi, K.; Ishizu, T.; Katagiri, D.; Fujita, T.; Matsubara, T.; Yahagi, N.; Sugaya, T.; Noiri, E. Evaluation of new acute kidney injury biomarkers in a mixed intensive care unit. Crit. Care Med. 2011, 39, 2464–2469. [Google Scholar] [CrossRef]
  87. Bennett, M.; Dent, C.L.; Ma, Q.; Dastrala, S.; Grenier, F.; Workman, R.; Syed, H.; Ali, S.; Barasch, J.; Devarajan, P. Urine NGAL predicts severity of acute kidney injury after cardiac surgery: A prospective study. Clin. J. Am. Soc. Nephrol. 2008, 3, 665–673. [Google Scholar] [CrossRef]
  88. Tian, M.; Zhao, S. The early diagnostic value of urinary Sema3A for ICU adult patients with acute kidney injury. Zhonghua Yi Xue Za Zhi 2015, 95, 1457–1462. [Google Scholar]
  89. Doi, K.; Noiri, E.; Nangaku, M.; Yahagi, N.; Jayakumar, C.; Ramesh, G. Repulsive guidance cue semaphorin 3A in urine predicts the progression of acute kidney injury in adult patients from a mixed intensive care unit. Nephrol. Dial. Transplant. 2014, 29, 73–80. [Google Scholar] [CrossRef]
  90. Fang, W.; Wang, Z.; Li, Q.; Wang, X.; Zhang, Y.; Sun, Y.; Tang, W.; Ma, C.; Sun, J.; Li, N.; et al. Gpr97 Exacerbates AKI by Mediating Sema3A Signaling. J. Am. Soc. Nephrol. 2018, 29, 1475–1489. [Google Scholar] [CrossRef]
  91. Kar, F.; Hacioglu, C.; Senturk, H.; Donmez, D.B.; Kanbak, G.; Uslu, S. Curcumin and LOXblock-1 ameliorate ischemia-reperfusion induced inflammation and acute kidney injury by suppressing the semaphorin-plexin pathway. Life Sci. 2020, 256, 118016. [Google Scholar] [CrossRef] [PubMed]
  92. Zhu, G.; Pei, L.; Lin, F.; Yin, H.; Li, X.; He, W.; Liu, N.; Gou, X. Exosomes from human-bone-marrow-derived mesenchymal stem cells protect against renal ischemia/reperfusion injury via transferring miR-199a-3p. J. Cell. Physiol. 2019, 234, 23736–23749. [Google Scholar] [CrossRef] [PubMed]
  93. Ruiz-Ortega, M.; Rayego-Mateos, S.; Lamas, S.; Ortiz, A.; Rodrigues-Diez, R.R. Targeting the progression of chronic kidney disease. Nat. Rev. Nephrol. 2020, 16, 269–288. [Google Scholar] [CrossRef] [PubMed]
  94. Viazzi, F.; Ramesh, G.; Jayakumar, C.; Leoncini, G.; Garneri, D.; Pontremoli, R. Increased urine semaphorin-3A is associated with renal damage in hypertensive patients with chronic kidney disease: A nested case-control study. J. Nephrol. 2015, 28, 315–320. [Google Scholar] [CrossRef] [PubMed]
  95. Iwano, M.; Plieth, D.; Danoff, T.M.; Xue, C.; Okada, H.; Neilson, E.G. Evidence that fibroblasts derive from epithelium during tissue fibrosis. J. Clin. Investig. 2002, 110, 341–350. [Google Scholar] [CrossRef]
  96. Takagawa, S.; Nakamura, F.; Kumagai, K.; Nagashima, Y.; Goshima, Y.; Saito, T. Decreased semaphorin3A expression correlates with disease activity and histological features of rheumatoid arthritis. BMC Musculoskelet. Disord. 2013, 14, 40. [Google Scholar] [CrossRef]
  97. Rimar, D.; Nov, Y.; Rosner, I.; Slobodin, G.; Rozenbaum, M.; Halasz, K.; Haj, T.; Jiries, N.; Kaly, L.; Boulman, N.; et al. Semaphorin 3A: An immunoregulator in systemic sclerosis. Rheumatol. Int. 2015, 35, 1625–1630. [Google Scholar] [CrossRef]
  98. Kiriakidou, M.; Ching, C.L. Systemic Lupus Erythematosus. Ann. Internet Med. 2020, 172, ITC81–ITC96. [Google Scholar] [CrossRef]
  99. Kessel, A.; Haj, T.; Peri, R.; Snir, A.; Melamed, D.; Sabo, E.; Toubi, E. Human CD19+CD25high B regulatory cells suppress proliferation of CD4+ T cells and enhance Foxp3 and CTLA-4 expression in T-regulatory cells. Autoimmun. Rev. 2012, 11, 670–677. [Google Scholar] [CrossRef]
  100. Luft, F.C. Semaphorin-3A is a repulsive but attractive renal guidance cue to therapy. J. Mol. Med. 2014, 92, 1225–1227. [Google Scholar] [CrossRef]
  101. Summers, S.A.; Hoi, A.; Steinmetz, O.M.; O’Sullivan, K.M.; Ooi, J.D.; Odobasic, D.; Akira, S.; Kitching, A.R.; Holdsworth, S.R. TLR9 and TLR4 are required for the development of autoimmunity and lupus nephritis in pristane nephropathy. J. Autoimmun. 2010, 35, 291–298. [Google Scholar] [CrossRef] [PubMed]
  102. Capolunghi, F.; Rosado, M.M.; Cascioli, S.; Girolami, E.; Bordasco, S.; Vivarelli, M.; Ruggiero, B.; Cortis, E.; Insalaco, A.; Fanto, N.; et al. Pharmacological inhibition of TLR9 activation blocks autoantibody production in human B cells from SLE patients. Rheumatology 2010, 49, 2281–2289. [Google Scholar] [CrossRef] [PubMed]
  103. Doron, R.; Merav, L.; Nasrin, E.; Adi, S.D.; Elias, T.; Gleb, S.; Itzhak, R.; Michael, R.; Zahava, V. Low Urine Secretion of Semaphorin3A in Lupus Patients with Proteinuria. Inflammation 2022, 45, 603–609. [Google Scholar] [CrossRef] [PubMed]
  104. Bejar, J.; Kessler, O.; Sabag, A.D.; Sabo, E.; Itzhak, O.B.; Neufeld, G.; Vadasz, Z. Semaphorin3A: A Potential Therapeutic Tool for Lupus Nephritis. Front. Immunol. 2018, 9, 634. [Google Scholar] [CrossRef]
  105. Florens, N. Cardiorenal Syndrome: Ebony and Ivory. Diagnostics 2023, 13, 1539. [Google Scholar] [CrossRef]
  106. Hu, H.; Xuan, Y.; Xue, M.; Cheng, W.; Wang, Y.; Li, X.; Yin, J.; Li, X.; Yang, N.; Shi, Y.; et al. Semaphorin 3A attenuates cardiac autonomic disorders and reduces inducible ventricular arrhythmias in rats with experimental myocardial infarction. BMC Cardiovasc. Disord. 2016, 16, 16. [Google Scholar] [CrossRef]
  107. Rienks, M.; Carai, P.; Bitsch, N.; Schellings, M.; Vanhaverbeke, M.; Verjans, J.; Cuijpers, I.; Heymans, S.; Papageorgiou, A. Sema3A promotes the resolution of cardiac inflammation after myocardial infarction. Basic Res. Cardiol. 2017, 112, 42. [Google Scholar] [CrossRef]
Figure 1. Structure of SEMA3A and its receptors, NRP1 and Plexin A. SEMA3A: semaphorin3A; SEMA3A-I: semaphorin3A inhibitor; NRP1: neuropilin-1; N: N-terminal; SEMA: SEMA domain; PSI: cysteine-rich plexin–semaphorin–integrin domain; Ig: immunoglobulin-like domain; Basic: basic domain; C: C-terminal; a1: complement binding domain a1; a2: complement binding domain a2; b1: coagulation factor V homology domain; b2: coagulation factor VIII homology domain; C: c-domain; IPT: Ig-like, plexin and transcription factor domain; GAP domain: GAP domain GTPase activating protein.
Figure 1. Structure of SEMA3A and its receptors, NRP1 and Plexin A. SEMA3A: semaphorin3A; SEMA3A-I: semaphorin3A inhibitor; NRP1: neuropilin-1; N: N-terminal; SEMA: SEMA domain; PSI: cysteine-rich plexin–semaphorin–integrin domain; Ig: immunoglobulin-like domain; Basic: basic domain; C: C-terminal; a1: complement binding domain a1; a2: complement binding domain a2; b1: coagulation factor V homology domain; b2: coagulation factor VIII homology domain; C: c-domain; IPT: Ig-like, plexin and transcription factor domain; GAP domain: GAP domain GTPase activating protein.
Diagnostics 13 03038 g001
Figure 2. SEMA3A and pathophysiology in kidney diseases. DN: diabetic nephropathy; CKD: chronic kidney disease; AKI: acute kidney injury; LN: lupus nephritis; EMT: epithelial–mesenchymal transition; SEMA3A: semaphorin3A; NRP1: neuropillin-1. Red up arrows: increase. Red down arrow: decrease.
Figure 2. SEMA3A and pathophysiology in kidney diseases. DN: diabetic nephropathy; CKD: chronic kidney disease; AKI: acute kidney injury; LN: lupus nephritis; EMT: epithelial–mesenchymal transition; SEMA3A: semaphorin3A; NRP1: neuropillin-1. Red up arrows: increase. Red down arrow: decrease.
Diagnostics 13 03038 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Sang, Y.; Tsuji, K.; Nakanoh, H.; Fukushima, K.; Kitamura, S.; Wada, J. Role of Semaphorin 3A in Kidney Development and Diseases. Diagnostics 2023, 13, 3038. https://doi.org/10.3390/diagnostics13193038

AMA Style

Sang Y, Tsuji K, Nakanoh H, Fukushima K, Kitamura S, Wada J. Role of Semaphorin 3A in Kidney Development and Diseases. Diagnostics. 2023; 13(19):3038. https://doi.org/10.3390/diagnostics13193038

Chicago/Turabian Style

Sang, Yizhen, Kenji Tsuji, Hiroyuki Nakanoh, Kazuhiko Fukushima, Shinji Kitamura, and Jun Wada. 2023. "Role of Semaphorin 3A in Kidney Development and Diseases" Diagnostics 13, no. 19: 3038. https://doi.org/10.3390/diagnostics13193038

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop