Sphingosine 1-phosphate in metabolic syndrome (Review)

  • Authors:
    • Wei Chen
    • Hongwei Lu
    • Jie Yang
    • Hong Xiang
    • Hui Peng
  • View Affiliations

  • Published online on: September 7, 2016     https://doi.org/10.3892/ijmm.2016.2731
  • Pages: 1030-1038
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Metabolic syndrome (MetS), a clustering of components, is closely associated with the development and prognosis of cardiovascular disease and diabetes. Sphingosine 1-phosphate (S1P) is a lysophospholipid with paracrine and autocrine effects, which is associated with obesity, insulin resistance, hyperglycemia, dyslipidemia and hypertension through extracellular and intracellular signals to achieve a variety of biological functions. However, there is controversy regarding the role of S1P in MetS; the specific role played by S1P remains unclear. It ameliorates abnormal energy metabolism and deviant adipogenesis and mediates inflammation in obesity. Despite the fact that sphingosine kinase (SphK)2/S1P increases the glucose‑stimulated insulin secretion of β-cells, more evidence showed that activation of the SphK1/S1P/S1P2R pathway inhibited the feedback loop of insulin secretion and sensitivity. The majority of S1P1R activation improves diabetes whereas S1P2R activation worsens the condition. In hyperlipidemia, S1P binds to high-density lipoprotein, low‑density lipoprotein and very low-density lipoprotein exerting different effects. Moreover, low concentrations of S1P lead to vasodilation whereas high concentrations of S1P result in vasocontraction of isolated arterioles. This review discusses the means by which different SphKs, S1P concentrations or S1P receptor subtypes results to diverse result in MetS, and then examines the role of S1P in MetS.

1. Introduction

Over the past decade, various diagnostic criteria for metabolic syndrome (MetS) have been proposed by different organizations (16). However, generally speaking, MetS is defined by a cluster of factors including obesity/central obesity, insulin resistance (IR), dyslipidemia and hypertension. As a worldwide problem, the prevalence of age-adjusted MetS estimated by the National Health and Nutrition Examination Survey (NHANES) is 22.9% in the US adult population (age ≥20) in the last decade (7). According to a representative sample of elderly people aged 60–95 in Beijing in two cross-sectional surveys, the prevalence of MetS in elderly Chinese individuals was 50.4% in 2001 and increased to 58.1% in 2010 (8). Besides high morbidity, MetS is associated with the development of cardiovascular disease (9), type 2 diabetes and cancer (10). Mottillo et al (11) found that MetS is associated with a 2-fold increase in cardiovascular outcomes. In addition, MetS is also involved in the development of cancer, as colorectal cancer is one of the most common types of cancer in patients with MetS (12).

Thus, it is important to identify a biomarker significantly associated with MetS. As a biological molecule involved in carbohydrate and lipid metabolism, sphingosine 1-phosphate (S1P) is a potent sphingolipid mediator that regulates an array of cellular responses, including cell migration, differentiation, apoptosis, lymphocyte trafficking and inflammation, by acting as both an extracellular signal and an intracellular second messenger (13). Herein, we reviewed the association between S1P and MetS, with emphasis upon the ways in which researchers have gained knowledge of this correlation.

2. S1P metabolism and signaling

Sphingosine, the substrate for S1P synthesis which is produced by the degradation of ceramide, may be phosphorylated by sphingosine kinases (SphKs) to form S1P. In subcellular organelles, S1P synthesis is initiated in the endoplasmic reticulum (ER), whereas later steps of the complex metabolism mainly occur in the Golgi apparatus, together with the involvement of lysosomes, nuclei and mitochondria (14). SphK1 is mainly localized in the cytoplasm and translocates to the cell membrane for activation, whereas SphK2 is primarily but not exclusively localized in the nucleus (15). S1P is either dephosphorylated by two S1P-specific phosphatases (SPP1 and SPP2), or irreversibly degraded by S1P lyase (SPL) to phosphoethanolamine (PE) and hexadecenal which are incorporated into glycerolipids. Extracellular S1P may also be degraded by lysophospholipid phosphatase (LPP)1 and LPP3 into sphingosine, which is taken up by cells for further metabolism (16).

In healthy subjects, the concentration of plasma S1P is 100–300 nM and approximately 100 nM in lymph (1721). Plasma S1P is principally derived from erythrocytes (22) and vascular endothelial cells (23), whereas the majority of lymph S1P originates from lymphatic endothelial cells (24). S1P in vascular and lymphatic endothelial cells is exported by the specific transporter Spns2 (25,26). However, in erythrocytes, S1P is exported by an ATP-dependent, vanadate- and glyburide-sensitive transporter. Requiring an extracellular stimulus such as thrombin, S1P is exported from platelets through two independent transporters, a Ca2+-dependent transporter and an ATP-dependent glyburide-sensitive transporter (27). The majority of plasma S1P is combined with apolipoprotein M (apoM) which preferentially associates with high-density lipoprotein (HDL) (28,29). The remaining part binds to albumin, low density lipoproteins (LDLs) and very low density lipoproteins (VLDLs) (29).

S1P signals through 5 specific G-coupled S1P receptors designated as S1P1R-5 (30,31) and each subtype exhibits differential coupling efficacy to G protein α subunits. Windh et al (32) revealed that S1P1R coupled exclusively to Gi, whereas both S1P2R and S1P4R coupled to Gi and to G12/13 as well as to the Gq family using the heterologous expression system of insect Sf9 cells. S1P4R (33) and S1P5R (34) were shown to couple to Gi and G12, by determining 35S-GTPγS binding to G proteins in CHO cells. This coupling resulted in the activation of small GTPases such as Rho, Rac and Ras (3537). Further downstream effectors of S1P receptors include adenylate cyclase, phosphatidylinositol 3-kinase (PI3K), phospholipase C, protein kinase C and intracellular calcium (38). Although widely expressed, S1P1R, S1P2R and S1P3R were principally expressed in vascular tissues, whereas S1P5R and S1P4R were largely expressed in the hematopoietic and nervous systems, respectively (39).

On the other hand, S1P has been suggested to function as an intracellular messenger. Parham et al found that S1P activated a luciferase-tagged peroxisome proliferator-activated receptor γ (PPARγ)-specific gene reporter by ~12-fold in human endothelial cells, independently of its known effects on canonical signaling through S1P receptors (40). S1P in the nucleus, produced by SphK2 bound to the histone deacetylases HDAC1 and HDAC2 specifically, inhibited their enzymatic activity, which then affects the dynamic balance of histone acetylation and thus, the epigenetic regulation of specific target genes (41). Similar to nuclear S1P, the majority of mitochondrial S1P produced by mitochondrial SphK2 specifically binds to homomeric PHB2, which regulates mitochondrial assembly and function. Furthermore, mitochondrial respiration through cytochrome oxidase was reduced by depleting SphK2 or PHB2, suggesting that the interaction between S1P and PHB2 is important for mitochondrial assembly and respiration (42). On the other hand, independently of S1P extracellular signals, SphK1 as well as the production of intracellular S1P are necessary for nuclear factor-κB (NF-κB) activation by tumor necrosis factor-α (TNF-α) (43) (Fig. 1).

Figure 1

Sphingosine 1-phosphate (S1P) metabolism and signaling. Sphingosine, the substrate for S1P synthesis, which is produced by the degradation of ceramide, may be phosphorylated by sphingosine kinases (SphK1 and SphK2) to form S1P. SphK1 is generally localized in the cytoplasm, whereas SphK2 is primarily but not exclusively localized in nucleus. In subcellular organelles, S1P synthesis initiates in the endoplasmic reticulum (ER), where S1P is irreversibly degraded by S1P lyase (SPL) to phosphoethanolamine (PE) or dephosphorylated by S1P phosphatases (SPPs) to sphingosine. In vascular and lymphatic endothelial cells, S1P is transported out by specific Spns2 transporters. S1P is exported out of vascular and lymphatic endothelial cells by the specific transporter Spns2. However, in erythrocytes, S1P is exported by an ATP-dependent, vanadate- and glyburide-sensitive transporter. The majority of plasma S1P is bound to apolipoprotein M (apoM) which preferentially associates with high densiy lipoprotein (HDL). The extracellular S1P signals through specific G-coupled S1P receptors designated as S1P1R-5, activating Rac, phosphatidylinositol 3-kinase (PI3K), adenylate cyclase (AC), extracellular-regulated protein kinase (ERK), phospholipase C (PLC), Rho and Jun N-terminal kinase (JNK) to contribute to cellular responses. Intracellular S1P also directly activates prohibitin 2 (PHB2) in the mitochondria, which stabilizes cytochrome c oxidase. In the nucleus, S1P is also involved in the activation of nuclear transcription factors PPARα and PPARγ, and histone deacetylases (HDACs).

3. S1P in obesity

S1P is positively associated with obesity. Plasma levels of S1P were higher in obese patients than those in non-obese and lean individuals (44). In addition, it was demonstrated that plasma S1P positively correlates with body mass index (BMI), total body fat percentage and waist circumference (45). The pathophysiology of obesity is complex; it is known to involve abnormal energy metabolism, deviant adipogenesis and inflammation. Recent studies have suggested that S1P is involved in these pathophysiological processes and in fact, reduces obesity.

S1P ameliorates obesity through the regulation of energy homeostasis. Silva et al (46) found that S1P1R protein is highly expressed in the hypothalamic pro-opiomelanocortin neurons of rats. Additionally, food consumption was decreased and energy expenditure was increased by intra-cerebroventricular injections of S1P through the activation of signal transducer and activator of transcription 3 (STAT3) and the melanocortin system, and vice versa. Moreover, the expression of S1P1R in neurons was controlled by STAT3 in neurons through a positive feedback mechanism. As several models of obesity display an imbalance of the hypothalamic S1P/S1P1R/STAT3 axis, whereas pharmacological intervention improves these phenotypes, this indicates that the S1P/S1P1R/STAT3 signaling pathway in neurons plays a vital role in controlling energy homeostasis in rats.

S1P reduces fat mass by affecting adipogenesis and lipolysis through several pathways. In maturing 3T3-L1 preadipocytes, S1P significantly decreased lipid accumulation in a dose-dependent manner with the downregulation of the transcriptional levels of the CCAAT/enhancer binding proteins α (C/EBPα), PPARγ and adiponectin, which are markers of adipogenic differentiation. Moreover, the activation of Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (p38 MAPK) were also downregulated by S1P treatment in human preadipocytes. These results suggest that S1P mediates the downregulation of adipogenic transcription factors and by inactivating the JNK and p38 MAPK signaling pathways subsequently affects adipogenesis (47). FTY720, an analogue of S1P, phosphorylated by SphK2 to form FTY720(S)-phosphate (FTY720-P), which binds to S1P1R, S1P3R, S1P4R and S1P5R, significantly down-regulated the markers of adipogenic differentiation (PPARγ, C/EBPα and adiponectin) and upregulated the regulators of lipolysis, [hormone-sensitive lipase (HSL), adipose triglyceride lipase (ATGL) and perilipin], indicating that FTY720 prevented obesity by modulating adipogenesis and lipolysis (48). This evidence suggests that S1P affects several pathways to adjust the balance between adipogenesis and lipolysis, subsequently altering the fat mass which reduces obesity, indicating that S1P may be used in the treatment of obesity. However, the internal mechanisms responsible for these effects remain to be elucidated. Further investigation is warranted into whether extracellular and intracellular S1P signals are involved in this process as well as into the mechanisms responsible for achieving these effects.

As chronic low-grade inflammation and activation of the immune system participate in the pathogenesis of MetS (49) inflammation is also involved in the interaction between S1P and obesity. A cross-sectional study recruited 30 healthy overweight and 15 lean adolescents, and found that there was a significant association between S1P and TNF-α (50). In animal studies, Samad et al found that S1P was elevated in obese (ob/ob) mice, and S1P induced the gene expression of interleukin (IL)-6, TNF-α, monocyte chemoattractant protein-1 and keratinocyte-derived chemokine in cultured adipocytes (51). On the other hand, Wang et al found that genetic disruption of the SphK1/S1P signaling pathway in mice with diet-induced obesity (DIO) increased adipose gene expression of the anti-inflammatory molecules IL-10 and adiponectin whereas it reduced adipose tissue macrophage recruitment and the expression of the proinflammatory molecules TNF-α and IL-6 (52). These findings suggest that the SK1/S1P signaling pathway contributed to the proinflammatory phenotype of the obese adipose tissue, and played a critical role in the pathogenesis of obesity-mediated metabolic disease.

As S1P ameliorates abnormal energy metabolism and deviant adipogenesis, while mediating inflammation, it plays an alternative role in the development of obesity. There is a possibility that, in the early stages of obesity, S1P increases the expression of proinflammatory factors, which activate the protective mechanisms of the body, thereby attenuating further development of obesity. Thus, S1P effectively adjusted the balance between adipogenesis and lipolysis in order to decrease the fat mass, which is the most prominent feature of obesity. On the other hand, S1P is a lipid with multiple biological activities mediated through intracellular and extracelluar pathways, and the activation of different pathways leads to different results. Thus, it is necessary to explore more specific mechanisms, and additional animal models and clinical trials are required in order to confirm the effects of S1P.

4. S1P in IR

S1P is synthesized by SphKs. SphK1 and SphK2 mediate opposite effects in insulin sensitivity. Qi et (53) found that all high fat diet (HFD)-fed SphK1(−/−) mice manifested evident diabetes, accompanied by a nearly 3-fold reduction in insulin levels compared with the WT mice which developed glucose intolerance and compensatory hyperinsulinemia. Pancreatic β-cell mass was increased by 140% in HFD-fed WT mice and decreased to 50% in HFD-fed SphK1(−/−) mice, in comparison with the chow diet control groups, respectively. The SphK1 level was also elevated in obese, type 2 diabetic humans. Enhanced insulin signaling in adipose and muscle as well as improved systemic insulin sensitivity and glucose tolerance were demonstrated in SphK1(−/−) mice, suggesting that the SphK1-S1P axis improves IR associated with obesity and type 2 diabetes (52). In addition, primary islets isolated from SphK1(−/−) mice exhibited higher susceptibility to lipotoxicity than WT controls. Of note, S1P profoundly abrogated lipotoxicity in β cells or the cells lacking SphK1 activity and SphK1(−/−) islets, highlighting a pivotal role of S1P in β-cell survival under lipotoxic conditions (53). Recognizing that S1P is metabolized by SphK2, the change of SphK2 is also involved in the process of IR. Cantrell Stanford et al (54) demonstrated that glucose elevates intracellular S1P by activating SphK2 in MIN6 cells and mouse pancreatic islets. Notably, the elevation of S1P correlates with the increase in glucose-stimulated insulin secretion (GSIS). On the other hand, downregulated levels of S1P and the knockdown of SphK2 in MIN6 cells or primary islets results in decreased GSIS, whereas the knockdown of the S1P phosphatase, SPP1, leads to a rise in GSIS. These data suggest that glucose-activated SphK2/S1P is important for GSIS in pancreatic β cells.

Apart from pancreatic islets, SphK1/S1P/S1P receptors also acted as an insulin-mimetic cue in insulin sensitive tissues. SphK1 transgenic mice fed an HFD showed increased SphK1 activity in skeletal muscle, accompanied by ameliorated muscle insulin resistance, which may be associated with a concomitant reduction in the phosphorylation of c-JNK, a serine threonine kinase associated with IR, thus indicting that skeletal muscle and whole-body insulin sensitivity were improved in SphK1 transgenic mice on an HFD (55). In addition, the inhibition of SphK1 decreased the effect of palmitate on insulin-stimulated glucose uptake by attenuating the activity of the AKT/glycogen synthase kinase 3β (GSK3β) signaling pathway and insulin signaling proteins in L6 myotubes (56) Furthermore, SphK1 gene delivery significantly enhanced the phosphorylation of insulin-signaling kinases such as Akt and GSK3β in the livers of diabetic animals (57). S1P, through engagement with S1P2R, has been found to produce a transient burst of reactive oxygen species (ROS) through calcium-dependent activation of the small GTPase Rac1 in skeletal muscle cells, accompanied by a redox modulation of protein tyrosine phosphatase-1B (TPT-1B) activity, the main negative regulator of insulin receptor phosphorylation. It is indicative of critical crosstalk occurring between S1P, insulin pathways and ROS (58). The extra- and intracellular S1P from the liver, another important organ involved in IR, was markedly increased by palmitate. Once generated, S1P bound to S1P2R via reduced insulin-mediated glycogen synthesis and PI3K/Akt-GSK3β activation to impair insulin signaling (59).

The malignant crosstalk of pancreatic islet β-cell dysfunction and insulin sensitivity leads to insulin resistance. In spite of the fact that SphK2/S1P increases the GSIS of β-cells, more evidence showed that SphK1/S1P/S1P2R pathway activation inhibits the feedback loop of insulin secretion and sensitivity.

5. S1P in hyperglycemia

Plasma levels of S1P in patients with type 2 diabetes were significantly higher (~50%) than those in healthy individuals (60). Additionally, plasma S1P positively correlated with HbA1c (%) levels in humans (45). It is well known that β-cell dysfunction and insulin resistance are the most important pathophysiological mechanisms associated with the development of diabetes. Zhao et al found that oral administration of FTY720 to diabetic (db/db) mice increased β-cell mass and blood insulin levels to normalize fasting blood glucose levels. Further investigation confirmed that S1P controls β-cell regeneration in the islets isolated from the treated mice by decreasing cyclin-dependent kinase inhibitor p57 (KIP2) levels and increasing cyclin D3 expression via the PI3K pathway (61).

More importantly, S1P ameliorates the complications of diabetes. Diabetic nephropathy (DN) is the leading cause of end-stage renal failure, which may be attenuated by S1P. FTY720 or SEW2871, a selective S1P1R agonist, significantly reduced urinary albumin excretion which led to tubule injury and increased urinary TNF-α in rats with streptozotocin (STZ)-induced diabeties (62). A dorsal skin wound was made in the healthy and diabetic mice; S1P injection alone promoted wound healing in the diabetic mice compared with the control, and the combination of S1P and JTE-013 (S1P2R antagonist) administration induced maximal wound healing in diabetic mice (63). These findings indicate that S1P possesses unique potential in the therapy of DN and diabetic wounds.

However, controversy still exists. As the connective tissue growth factor (CTGF) is a marker in mesangial cells for the progression of DN, El-Shewy et al found that SphK1/S1P1/3R activation is upstream of extracellular regulated protein kinases (ERK)1/2 and JNK, which are essential for LDL-regulated expression of CTGF in renal mesangial cells (64). The SphK1/S1P pathway also plays a critical role in fibronectin accumulation, which is a glomerular extracellular matrix protein associated with DN, in both kidneys from STZ-induced diabetic rats and high glucose-treated glomerular mesangial cells (65). Lan et al (66) identified that fibronectin expression was upregulated via S1P-S1P2R-MAPK (ERK1/2 and p38 MAPK) axis activation in mesangial cells under high glucose conditions, suggesting that SphK1-S1P-S1P2R-MAPK may be a potential therapeutic target for the treatment of DN.

In diabetes, elevated S1P levels are evident. While the downstream receptors have the opposite effect under hyper-glycemic conditions, S1P1R activation ameliorates diabetes whereas S1P2R activation worsens the condition (Table I). Finding an equilibrium point between S1P1R and S1P2R, and then balancing the activation of S1P1R and S1P2R, may provide a novel target for the treatment of diabetes.

Table I

Role of S1P and downstream signaling in insulin resistance and hyperglycemia.

Table I

Role of S1P and downstream signaling in insulin resistance and hyperglycemia.

SubjectsIntervening substance/target moleculesDownstream signalBiological outcomeRefs.
SphK1 transgenic miceFed an HFDA concomitant reduction in the phosphorylation of c-JNKAmeliorate muscle insulin resistance(55)
MIN6 cells, pancreatic isletsGlucoseActivate SphK2/S1PIncreased GSIS(54)
db/db miceFTY720 (10 mg/kg, intraperitoneal injection daily for 6 weeks)Increase cyclin D3 and decline KIP2 via PI3KIncrease β-cell mass and blood insulin levels(61)
STZ-exposed diabetic ratsFTY720 (0.3 mg/kg, oral gavage, 9 weeks)Upregulate S1P1RReduce the urinary albumin excretion(62)
db/db miceS1P (10 or 100 mM injected into the wound bed daily) and JTE-013Downregulate S1P2RPromote the healing of wound(63)
DIO miceSphK1 deficiencyIncrease IL-10 and adiponectin levels; reduce ATM recruitment, TNF and IL-6Enhanced insulin signaling in adipose and muscle and improved systemic insulin sensitivity and glucose tolerance(52)
C2C12 cellsS1P (0.01–5 µM, 15–90 min)Calcium-dependent activation of the small GTPase Rac1Produce a transient burst of ROS(58)
Primary rat hepatocytesS1P (1 Mm, 5 h)S1PR2, reduces PI3K/Akt-GSK-3β activationReduce insulin mediated glycogen synthesis and impair insulin signaling(59)
Renal mesangial cellsSphKs inhibitor, VPC23019Inhibit activation of ERK1/2 and JNK via S1PR1 and S1P3REssential for expression of CTGF induced by LDL(64)
SphK1, S1P (1 µm, 12 h)S1P2R-MAPK (ERK1/2 and p38 MAPK)Mediate high glucose-induced fibronectin expression(66)

[i] HFD, high fat diet; c-JNK, c-Jun N-terminal kinase; GSIS, glucose-stimulated insulin secretion; KIP2, cyclin-dependent kinase inhibitor p57; PI3K, phosphatidylinositol 3-kinase; JTE-013, S1P2R antagonist; ATM, adipose tissue macrophage; ROS, reactive oxygen species; VPC23019, S1P3R antagonist; AKT, protein kinase B; ERK, extracellular regulated protein kinases; GSK3α/β, glycogen synthase kinase 3α/β; CTGF, connective tissue growth factor; p38 MAPK, p38 mitogen-activated protein kinase.

6. S1P in hyperlipidemia

S1P contributes to the protective role of HDL. Tong et al (67) found that HDL in diabetic patients with elevated S1P levels exhibited more powerful protective effects, inducing COX-2 expression and PGI2 release from human umbilical vein endothelial cells, than control HDL through the activation of S1P1R/3R. Additionally, S1P compensated for the reduction in COX-2 expression through glycated HDL by phosphorylating the ERK/MAPK-CREB pathway (68). These findings confirmed the protective role of HDL-binding S1P in patients with type 2 diabetes mellitus, suggesting a possible novel therapeutic target. There is no S1P in HDL in apoM(−/−) mice, whereas HDL in transgenic mice overexpressing human apoM has an increased S1P content, indicating that the bridge between HDL and S1P in plasma is apoM. The 1.7-Å structure of the S1P-apoM complex suggests that S1P specifically interacts with an amphiphilic pocket in the lipocalin fold of apoM. Human apoM(+) HDL induced S1P1R internalization, downstream MAPK and Akt activation, endothelial cell migration, and formation of endothelial adherence junctions, whereas apoM(−) HDL did not. Moreover, the HDL fraction of apoM(−/−) mice which lacked S1P showed dampened basal endothelial barrier function in lung tissue. This demonstrated that apoM is an essential vascular protective constituent of HDL by delivering S1P to the S1P1R on endothelial cells (28).

Plasma S1P not only positively correlated with LDL cholesterol in a human study (45), but also in experimental animal studies, showing the important role of S1P in the context of hyperlipidemia. High fructose-fed rats induced hyperlipidemia and activated the SphK1/S1P signaling pathway, which in turn led to the activation of NF-κB signaling and inflammation in rat livers. Importantly, lipid metabolic disorder as well as hepatic insulin and leptin signaling impairment were observed in this animal model, which resulted in lipid accumulation in the liver (69). Additionally, SphK1 gene delivery significantly attenuated elevated levels of plasma non-esterified fatty acid (NEFA), triacylglycerol, LDL and cholesterol, which markedly ameliorated liver, heart and kidney injuries induced by hyperglycaemia in KK/Ay diabetic mice (57). As in other animal models of hyperlipidemia, the capability to generate and release S1P were significantly higher in the hypersensitized platelets and blood plasma obtained from rabbits with hypercholesterolemia. Moreover, co-treatment with S1P (0.125–0.5 mM) potentiated the ox-LDL-induced proliferation of vascular smooth muscle cells in a dose-dependent manner through the S1P1R dependent pathway (70). Thus, the SphK1/S1P/S1P receptor signaling pathway may be a novel mechanism which is important in hyperlipidemia.

It is well known that hyperlipidemia is always accompanied by a low HDL level, and the majority of plasma S1P is bound to HDL. It has been hypothesized that amount of S1P which binds to HDL is less, whereas more S1P may bind to LDL and VLDL under conditions of hyperlipidemia. From the above studies, we can deduce that S1P exhibits a protective function by binding HDL, wheras the combination with LDL shows the opposite effects.

7. S1P in hypertension

Plasma S1P levels are significantly higher in patients with hypertension in comparison with normotensive patients. In fact, the S1P1R gene is a candidate for the control of salt sensitivity and hypertension in the stroke-prone spontaneously hypertensive rat (SHRSP) (71). In SHR, application of dimethylsphingosine, a sphingosine kinase inhibitor, led to a marked upregulation in mean arterial pressure (MAP) with a further rise in carotid artery resistance. However, in Wistar-Kyoto (WKY) rats, dimethylsphingosine had little effect on MAP (72). S1P induced transient relaxation of isolated pressurized mesenteric arterioles with low concentrations (10–100 nM) in a dose dependent manner. Maximal vasodilation (55±8%) was demonstrated at 2 min after S1P addition and returned to baseline levels by 5 min via G protein-dependent, calcium-sensitive, and PI3K signaling pathways (73).

However, controversy continues to exist. FTY720 produced modest hypertension (2–3 mmHg) in patients in a 1 year trial. Furthermore, FTY720 elicited dose-dependent hypertension after multiple days of oral administration through the activation of S1P3R in rats (74). Furthermore, S1P induced vasocontraction through the activation of the Rho/Rho-kinase signaling pathway in the canine basilar artery at high concentrations ranging between 100 and 10 mM (75). The S1P extracellular signals induced hypertension. S1P/S1P3R selectively constricted isolated cerebral arteries mainly by Rho and partly through G(i/o) protein (76). On the other hand, Yogi et al (77) identified that S1P/S1P1R is associated with specific pathways through epidermal growth factor receptor and platelet-derived growth factor transactivation, a process upregulated in the vascular smooth muscle cells of SHRSP rats which contributes to vascular inflammation in hypertension.

The complex cardiovascular effects of S1P are likely to be due to differential concentrations of S1P. Low concentrations (10–100 nM) lead to vasodilation whereas high concentrations (100–10,000 nM) result in the vasocontraction of isolated arterioles. The activation of different S1P receptor subtypes may also contribute to different results. Thus, the investigation of more mechanisms is necessary in order to identify the multiple roles of S1P in hypertension.

8. Conclusion

Due to the high prevalence of MetS worldwide, a condition which is closely assicated with the development and prognosis of diabetes and cardiovascular disease, studies were taken to find a biomarker or effective way to diminish the development and progress of MetS. S1P has been demonstrated to play multiple roles in the occurrence and development of this disease, mainly through S1P1R-S1P3R. There is controversy regarding the role of S1P in MetS, such as anti-inflammation versus pro-inflammation, and anti-hypertension versus pro-hypertension. Different SphKs, S1P concentration or S1P receptor subtypes result in diverse results (Fig. 2). Thus, the possible implications of S1P-directed mechanisms on the occurrence and development of MetS remains to be elucidated. Further investigations may provide novel candidates for the treatment of MetS.

Acknowledgments

The present study was supported by funding from the National Natural Science Foundation of China (81470593), the National Basic Research Program of China (2014CB542400) and the National Natural Science Foundation of China (81300053), the Fundamental Research Funds for the Central Universities of Central South University (2015zzts123).

References

1 

Alberti KG and Zimmet PZ: Definition, diagnosis and classification of diabetes mellitus and its complications. Part 1: diagnosis and classification of diabetes mellitus provisional report of a WHO consultation. Diabet Med. 15:539–553. 1998. View Article : Google Scholar : PubMed/NCBI

2 

Balkau B and Charles MA: Comment on the provisional report from the WHO consultation. European Group for the Study of Insulin Resistance (EGIR). Diabet Med. 16:442–443. 1999. View Article : Google Scholar : PubMed/NCBI

3 

Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults: Executive Summary of The Third Report of The National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, And Treatment of High Blood Cholesterol In Adults (Adult Treatment Panel III). JAMA. 285:2486–2497. 2001. View Article : Google Scholar : PubMed/NCBI

4 

Bloomgarden ZT: American Association of Clinical Endocrinologists (AACE) consensus conference on the insulin resistance syndrome: 25–26 August 2002, Washington, DC. Diabetes Care. 26:933–939. 2003. View Article : Google Scholar : PubMed/NCBI

5 

Alberti KG and Zimmet P: The metabolic syndrome - a new worldwide definition. Lancet. 366:1059–1062. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Alberti KG, Eckel RH, Grundy SM, Zimmet PZ, Cleeman JI, Donato KA, Fruchart JC, James WP, Loria CM and Smith SC Jr; International Diabetes Federation Task Force on Epidemiology and Prevention; Hational Heart, Lung, and Blood Institute; American Heart Association; World Heart Federation; International Atherosclerosis Society; International Association for the Study of Obesity: Harmonizing the metabolic syndrome: a joint interim statement of the International Diabetes Federation Task Force on Epidemiology and Prevention; National Heart, Lung, and Blood Institute; American Heart Association; World Heart Federation; International Atherosclerosis Society; and International Association for the Study of Obesity. Circulation. 120:1640–1645. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Beltrán-Sánchez H, Harhay MO, Harhay MM and McElligott S: Prevalence and trends of metabolic syndrome in the adult U.S. population, 1999–2010. J Am Coll Cardiol. 62:697–703. 2013. View Article : Google Scholar

8 

Liu M, Wang J, Jiang B, Sun D, Wu L, Yang S, Wang Y, Li X and He Y: Increasing prevalence of metabolic syndrome in a Chinese elderly population: 2001–2010. PLoS One. 8:e662332013. View Article : Google Scholar

9 

Wilson PW, D'Agostino RB, Parise H, Sullivan L and Meigs JB: Metabolic syndrome as a precursor of cardiovascular disease and type 2 diabetes mellitus. Circulation. 112:3066–3072. 2005. View Article : Google Scholar : PubMed/NCBI

10 

Mendonça FM, de Sousa FR, Barbosa AL, Martins SC, Araújo RL, Soares R and Abreu C: Metabolic syndrome and risk of cancer: Which link? Metabolism. 64:182–189. 2015. View Article : Google Scholar

11 

Mottillo S, Filion KB, Genest J, Joseph L, Pilote L, Poirier P, Rinfret S, Schiffrin EL and Eisenberg MJ: The metabolic syndrome and cardiovascular risk a systematic review and meta-analysis. J Am Coll Cardiol. 56:1113–1132. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Esposito K, Chiodini P, Capuano A, Bellastella G, Maiorino MI, Rafaniello C, Panagiotakos DB and Giugliano D: Colorectal cancer association with metabolic syndrome and its components: a systematic review with meta-analysis. Endocrine. 44:634–647. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Blaho VA and Hla T: Regulation of mammalian physiology, development, and disease by the sphingosine 1-phosphate and lysophosphatidic acid receptors. Chem Rev. 111:6299–6320. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Maceyka M and Spiegel S: Sphingolipid metabolites in inflammatory disease. Nature. 510:58–67. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Chan H and Pitson SM: Post-translational regulation of sphingosine kinases. Biochim Biophys Acta. 1831:147–156. 2013. View Article : Google Scholar

16 

Escalante-Alcalde D, Hernandez L, Le Stunff H, Maeda R, Lee HS, Gang-Cheng Jr, Sciorra VA, Daar I, Spiegel S, Morris AJ and Stewart CL: The lipid phosphatase LPP3 regulates extra-embryonic vasculogenesis and axis patterning. Development. 130:4623–4637. 2003. View Article : Google Scholar : PubMed/NCBI

17 

Knapp M, Lisowska A, Zabielski P, Musiał W and Baranowski M: Sustained decrease in plasma sphingosine-1-phosphate concentration and its accumulation in blood cells in acute myocardial infarction. Prostaglandins Other Lipid Mediat. 106:53–61. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Baranowski M, Charmas M, Długołęcka B and Górski J: Exercise increases plasma levels of sphingoid base-1 phosphates in humans. Acta Physiol (Oxf). 203:373–380. 2011. View Article : Google Scholar

19 

Knapp M, Baranowski M, Lisowska A and Musiał W: Decreased free sphingoid base concentration in the plasma of patients with chronic systolic heart failure. Adv Med Sci. 57:100–105. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Knapp M, Lisowska A, Knapp P and Baranowski M: Dose-dependent effect of aspirin on the level of sphingolipids in human blood. Adv Med Sci. 58:274–281. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Baranowski M, Górski J, Klapcinska B, Waskiewicz Z and Sadowska-Krepa E: Ultramarathon run markedly reduces plasma sphingosine-1-phosphate concentration. Int J Sport Nutr Exerc Metab. 24:148–156. 2014. View Article : Google Scholar

22 

Pappu R, Schwab SR, Cornelissen I, Pereira JP, Regard JB, Xu Y, Camerer E, Zheng YW, Huang Y, Cyster JG and Coughlin SR: Promotion of lymphocyte egress into blood and lymph by distinct sources of sphingosine-1-phosphate. Science. 316:295–298. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Venkataraman K, Lee YM, Michaud J, Thangada S, Ai Y, Bonkovsky HL, Parikh NS, Habrukowich C and Hla T: Vascular endothelium as a contributor of plasma sphingosine 1-phosphate. Circ Res. 102:669–676. 2008. View Article : Google Scholar : PubMed/NCBI

24 

Pham TH, Baluk P, Xu Y, Grigorova I, Bankovich AJ, Pappu R, Coughlin SR, McDonald DM, Schwab SR and Cyster JG: Lymphatic endothelial cell sphingosine kinase activity is required for lymphocyte egress and lymphatic patterning. J Exp Med. 207:17–27. 2010. View Article : Google Scholar :

25 

Mendoza A, Bréart B, Ramos-Perez WD, Pitt LA, Gobert M, Sunkara M, Lafaille JJ, Morris AJ and Schwab SR: The transporter Spns2 is required for secretion of lymph but not plasma sphingosine-1-phosphate. Cell Rep. 2:1104–1110. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Fukuhara S, Simmons S, Kawamura S, Inoue A, Orba Y, Tokudome T, Sunden Y, Arai Y, Moriwaki K, Ishida J, et al: The sphingosine-1-phosphate transporter Spns2 expressed on endothelial cells regulates lymphocyte trafficking in mice. J Clin Invest. 122:1416–1426. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Kobayashi N, Kobayashi N, Yamaguchi A and Nishi T: Characterization of the ATP-dependent sphingosine 1-phosphate transporter in rat erythrocytes. J Biol Chem. 284:21192–21200. 2009. View Article : Google Scholar : PubMed/NCBI

28 

Christoffersen C, Obinata H, Kumaraswamy SB, Galvani S, Ahnström J, Sevvana M, Egerer-Sieber C, Muller YA, Hla T, Nielsen LB and Dahlbäck B: Endothelium-protective sphingosine-1-phosphate provided by HDL-associated apolipoprotein M. Proc Natl Acad Sci USA. 108:9613–9618. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Okajima F: Plasma lipoproteins behave as carriers of extracellular sphingosine 1-phosphate: is this an atherogenic mediator or an antiatherogenic mediator? Biochim Biophys Acta. 1582:132–137. 2002. View Article : Google Scholar : PubMed/NCBI

30 

Lee MJ, Van Brocklyn JR, Thangada S, Liu CH, Hand AR, Menzeleev R, Spiegel S and Hla T: Sphingosine-1-phosphate as a ligand for the G protein-coupled receptor EDG-1. Science. 279:1552–1555. 1998. View Article : Google Scholar : PubMed/NCBI

31 

Rosen H, Stevens RC, Hanson M, Roberts E and Oldstone MB: Sphingosine-1-phosphate and its receptors: structure, signaling, and influence. Annu Rev Biochem. 82:637–662. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Windh RT, Lee MJ, Hla T, An S, Barr AJ and Manning DR: Differential coupling of the sphingosine 1-phosphate receptors Edg-1, Edg-3, and H218/Edg-5 to the G(i), G(q), and G(12) families of heterotrimeric G proteins. J Biol Chem. 274:27351–27358. 1999. View Article : Google Scholar : PubMed/NCBI

33 

Yamazaki Y, Kon J, Sato K, Tomura H, Sato M, Yoneya T, Okazaki H, Okajima F and Ohta H: Edg-6 as a putative sphingosine 1-phosphate receptor coupling to Ca(2+) signaling pathway. Biochem Biophys Res Commun. 268:583–589. 2000. View Article : Google Scholar : PubMed/NCBI

34 

Im DS, Heise CE, Ancellin N, O'Dowd BF, Shei GJ, Heavens RP, Rigby MR, Hla T, Mandala S, McAllister G, et al: Characterization of a novel sphingosine 1-phosphate receptor, Edg-8. J Biol Chem. 275:14281–14286. 2000. View Article : Google Scholar : PubMed/NCBI

35 

Ye D and Lin F: S1pr2/Gα13 signaling controls myocardial migration by regulating endoderm convergence. Development. 140:789–799. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Singleton PA, Dudek SM, Chiang ET and Garcia JG: Regulation of sphingosine 1-phosphate-induced endothelial cytoskeletal rearrangement and barrier enhancement by S1P1 receptor, PI3 kinase, Tiam1/Rac1, and alpha-actinin. FASEB J. 19:1646–1656. 2005. View Article : Google Scholar : PubMed/NCBI

37 

Ishimaru N, Yamada A, Nitta T, Arakaki R, Lipp M, Takahama Y and Hayashi Y: CCR7 with S1P1 signaling through AP-1 for migration of Foxp3+ regulatory T-cells controls autoimmune exocrinopathy. Am J Pathol. 180:199–208. 2012. View Article : Google Scholar

38 

Mendelson K, Evans T and Hla T: Sphingosine 1-phosphate signalling. Development. 141:5–9. 2014. View Article : Google Scholar :

39 

Waeber C; Sphingosine 1-phosphate (S1P) signaling and the vasculature: Lysophospholipid Receptors: Signaling and Biochemistry. Chun J, Hla T, Spiegel S and Moolenaar W: John Wiley and Sons, Inc; Hoboken, NJ: pp. 313–347. 2013, View Article : Google Scholar

40 

Parham KA, Zebol JR, Tooley KL, Sun WY, Moldenhauer LM, Cockshell MP, Gliddon BL, Moretti PA, Tigyi G, Pitson SM and Bonder CS: Sphingosine 1-phosphate is a ligand for peroxisome proliferator-activated receptor-γ that regulates neoangiogenesis. FASEB J. 29:3638–3653. 2015. View Article : Google Scholar : PubMed/NCBI

41 

Hait NC, Allegood J, Maceyka M, Strub GM, Harikumar KB, Singh SK, Luo C, Marmorstein R, Kordula T, Milstien S and Spiegel S: Regulation of histone acetylation in the nucleus by sphingosine-1-phosphate. Science. 325:1254–1257. 2009. View Article : Google Scholar : PubMed/NCBI

42 

Strub GM, Paillard M, Liang J, Gomez L, Allegood JC, Hait NC, Maceyka M, Price MM, Chen Q, Simpson DC, et al: Sphingosine-1-phosphate produced by sphingosine kinase 2 in mitochondria interacts with prohibitin 2 to regulate complex IV assembly and respiration. FASEB J. 25:600–612. 2011. View Article : Google Scholar :

43 

Alvarez SE, Harikumar KB, Hait NC, Allegood J, Strub GM, Kim EY, Maceyka M, Jiang H, Luo C, Kordula T, et al: Sphingosine-1-phosphate is a missing cofactor for the E3 ubiquitin ligase TRAF2. Nature. 465:1084–1088. 2010. View Article : Google Scholar : PubMed/NCBI

44 

Ito S, Iwaki S, Koike K, Yuda Y, Nagasaki A, Ohkawa R, Yatomi Y, Furumoto T, Tsutsui H, Sobel BE and Fujii S: Increased plasma sphingosine-1-phosphate in obese individuals and its capacity to increase the expression of plasminogen activator inhibitor-1 in adipocytes. Coron Artery Dis. 24:642–650. 2013.PubMed/NCBI

45 

Kowalski GM, Carey AL, Selathurai A, Kingwell BA and Bruce CR: Plasma sphingosine-1-phosphate is elevated in obesity. PLoS One. 8:e724492013. View Article : Google Scholar : PubMed/NCBI

46 

Silva VR, Micheletti TO, Pimentel GD, Katashima CK, Lenhare L, Morari J, Mendes MC, Razolli DS, Rocha GZ, de Souza CT, et al: Hypothalamic S1P/S1PR1 axis controls energy homeostasis. Nat Commun. 5:48592014. View Article : Google Scholar : PubMed/NCBI

47 

Moon MH, Jeong JK, Lee YJ, Seol JW and Park SY: Sphingosine-1-phosphate inhibits the adipogenic differentiation of 3T3-L1 preadipocytes. Int J Mol Med. 34:1153–1158. 2014.PubMed/NCBI

48 

Moon MH, Jeong JK, Lee JH, Park YG, Lee YJ, Seol JW and Park SY: Antiobesity activity of a sphingosine 1-phosphate analogue FTY720 observed in adipocytes and obese mouse model. Exp Mol Med. 44:603–614. 2012. View Article : Google Scholar : PubMed/NCBI

49 

Esser N, Legrand-Poels S, Piette J, Scheen AJ and Paquot N: Inflammation as a link between obesity, metabolic syndrome and type 2 diabetes. Diabetes Res Clin Pract. 105:141–150. 2014. View Article : Google Scholar : PubMed/NCBI

50 

Majumdar I and Mastrandrea LD: Serum sphingolipids and inflammatory mediators in adolescents at risk for metabolic syndrome. Endocrine. 41:442–449. 2012. View Article : Google Scholar : PubMed/NCBI

51 

Samad F, Hester KD, Yang G, Hannun YA and Bielawski J: Altered adipose and plasma sphingolipid metabolism in obesity: a potential mechanism for cardiovascular and metabolic risk. Diabetes. 55:2579–2587. 2006. View Article : Google Scholar : PubMed/NCBI

52 

Wang J, Badeanlou L, Bielawski J, Ciaraldi TP and Samad F: Sphingosine kinase 1 regulates adipose proinflammatory responses and insulin resistance. Am J Physiol Endocrinol Metab. 306:E756–E768. 2014. View Article : Google Scholar : PubMed/NCBI

53 

Qi Y, Chen J, Lay A, Don A, Vadas M and Xia P: Loss of sphingosine kinase 1 predisposes to the onset of diabetes via promoting pancreatic β-cell death in diet-induced obese mice. FASEB J. 27:4294–4304. 2013. View Article : Google Scholar : PubMed/NCBI

54 

Cantrell Stanford J, Morris AJ, Sunkara M, Popa GJ, Larson KL and Özcan S: Sphingosine 1-phosphate (S1P) regulates glucose-stimulated insulin secretion in pancreatic beta cells. J Biol Chem. 287:13457–13464. 2012. View Article : Google Scholar : PubMed/NCBI

55 

Bruce CR, Risis S, Babb JR, Yang C, Kowalski GM, Selathurai A, Lee-Young RS, Weir JM, Yoshioka K, Takuwa Y, et al: Overexpression of sphingosine kinase 1 prevents ceramide accumulation and ameliorates muscle insulin resistance in high-fat diet-fed mice. Diabetes. 61:3148–3155. 2012. View Article : Google Scholar : PubMed/NCBI

56 

Mikłosz A, Łukaszuk B, Baranowski M, Górski J and Chabowski A: Effects of inhibition of serine palmitoyltransferase (SPT) and sphingosine kinase 1 (SphK1) on palmitate induced insulin resistance in L6 myotubes. PLoS One. 8:e855472013. View Article : Google Scholar

57 

Ma MM, Chen JL, Wang GG, Wang H, Lu Y, Li JF, Yi J, Yuan YJ, Zhang QW, Mi J, et al: Sphingosine kinase 1 participates in insulin signalling and regulates glucose metabolism and homeostasis in KK/Ay diabetic mice. Diabetologia. 50:891–900. 2007. View Article : Google Scholar : PubMed/NCBI

58 

Rapizzi E, Taddei ML, Fiaschi T, Donati C, Bruni P and Chiarugi P: Sphingosine 1-phosphate increases glucose uptake through trans-activation of insulin receptor. Cell Mol Life Sci. 66:3207–3218. 2009. View Article : Google Scholar : PubMed/NCBI

59 

Fayyaz S, Henkel J, Japtok L, Krämer S, Damm G, Seehofer D, Püschel GP and Kleuser B: Involvement of sphingosine 1-phosphate in palmitate-induced insulin resistance of hepatocytes via the S1P2 receptor subtype. Diabetologia. 57:373–382. 2014. View Article : Google Scholar

60 

Randriamboavonjy V, Badenhoop K, Schmidt H, Geisslinger G, Fisslthaler B and Fleming I: The S1P(2) receptor expressed in human platelets is linked to the RhoA-Rho kinase pathway and is down regulated in type 2 diabetes. Basic Res Cardiol. 104:333–340. 2009. View Article : Google Scholar : PubMed/NCBI

61 

Zhao Z, Choi J, Zhao C and Ma ZA: FTY720 normalizes hyperglycemia by stimulating β-cell in vivo regeneration in db/db mice through regulation of cyclin D3 and p57(KIP2). J Biol Chem. 287:5562–5573. 2012. View Article : Google Scholar

62 

Awad AS, Rouse MD, Khutsishvili K, Huang L, Bolton WK, Lynch KR and Okusa MD: Chronic sphingosine 1-phosphate 1 receptor activation attenuates early-stage diabetic nephropathy independent of lymphocytes. Kidney Int. 79:1090–1098. 2011. View Article : Google Scholar : PubMed/NCBI

63 

Kawanabe T, Kawakami T, Yatomi Y, Shimada S and Soma Y: Sphingosine 1-phosphate accelerates wound healing in diabetic mice. J Dermatol Sci. 48:53–60. 2007. View Article : Google Scholar : PubMed/NCBI

64 

El-Shewy HM, Sohn M, Wilson P, Lee MH, Hammad SM, Luttrell LM and Jaffa AA: Low-density lipoprotein induced expression of connective tissue growth factor via transactivation of sphingosine 1-phosphate receptors in mesangial cells. Mol Endocrinol. 26:833–845. 2012. View Article : Google Scholar : PubMed/NCBI

65 

Liu W, Lan T, Xie X, Huang K, Peng J, Huang J, Shen X, Liu P and Huang H: S1P2 receptor mediates sphingosine-1-phosphate-induced fibronectin expression via MAPK signaling pathway in mesangial cells under high glucose condition. Exp Cell Res. 318:936–943. 2012. View Article : Google Scholar : PubMed/NCBI

66 

Lan T, Liu W, Xie X, Xu S, Huang K, Peng J, Shen X, Liu P, Wang L, Xia P and Huang H: Sphingosine kinase-1 pathway mediates high glucose-induced fibronectin expression in glomerular mesangial cells. Mol Endocrinol. 25:2094–2105. 2011. View Article : Google Scholar : PubMed/NCBI

67 

Tong X, Peng H, Liu D, Ji L, Niu C, Ren J, Pan B, Hu J, Zheng L and Huang Y: High-density lipoprotein of patients with type 2 diabetes mellitus upregulates cyclooxgenase-2 expression and prostacyclin I-2 release in endothelial cells: relationship with HDL-associated sphingosine-1-phosphate. Cardiovasc Diabetol. 12:272013. View Article : Google Scholar : PubMed/NCBI

68 

Tong X, Lv P, Mathew AV, Liu D, Niu C, Wang Y, Ji L, Li J, Fu Z, Pan B, et al: The compensatory enrichment of sphingosine-1-phosphate harbored on glycated high-density lipoprotein restores endothelial protective function in type 2 diabetes mellitus. Cardiovasc Diabetol. 13:822014. View Article : Google Scholar

69 

Wang X, Zhang DM, Gu TT, Ding XQ, Fan CY, Zhu Q, Shi YW, Hong Y and Kong LD: Morin reduces hepatic inflammation-associated lipid accumulation in high fructose-fed rats via inhibiting sphingosine kinase 1/sphingosine 1-phosphate signaling pathway. Biochem Pharmacol. 86:1791–1804. 2013. View Article : Google Scholar : PubMed/NCBI

70 

Son DJ, Lee HW, Shin HW, Lee JJ, Yoo HS, Kim TJ, Yun YP and Hong JT: Enhanced release of sphingosine-1-phosphate from hypercholesterolemic platelets: role in development of hypercholesterolemic atherosclerosis. Prostaglandins Leukot Essent Fatty Acids. 78:383–390. 2008. View Article : Google Scholar : PubMed/NCBI

71 

Graham D, McBride MW, Gaasenbeek M, Gilday K, Beattie E, Miller WH, McClure JD, Polke JM, Montezano A, Touyz RM and Dominiczak AF: Candidate genes that determine response to salt in the stroke-prone spontaneously hypertensive rat: congenic analysis. Hypertension. 50:1134–1141. 2007. View Article : Google Scholar : PubMed/NCBI

72 

Spijkers LJ, van den Akker RF, Janssen BJ, Debets JJ, De Mey JG, Stroes ES, van den Born BJ, Wijesinghe DS, Chalfant CE, MacAleese L, et al: Hypertension is associated with marked alterations in sphingolipid biology: a potential role for ceramide. PLoS One. 6:e218172011. View Article : Google Scholar : PubMed/NCBI

73 

Dantas AP, Igarashi J and Michel T: Sphingosine 1-phosphate and control of vascular tone. Am J Physiol Heart Circ Physiol. 284:H2045–H2052. 2003. View Article : Google Scholar : PubMed/NCBI

74 

Fryer RM, Muthukumarana A, Harrison PC, Nodop Mazurek S, Chen RR, Harrington KE, Dinallo RM, Horan JC, Patnaude L, Modis LK and Reinhart GA: The clinically-tested S1P receptor agonists, FTY720 and BAF312, demonstrate subtype-specific bradycardia (S1P1) and hypertension (S1P3) in rat. PLoS One. 7:e529852012. View Article : Google Scholar

75 

Tosaka M, Okajima F, Hashiba Y, Saito N, Nagano T, Watanabe T, Kimura T and Sasaki T: Sphingosine 1-phosphate contracts canine basilar arteries in vitro and in vivo: possible role in pathogenesis of cerebral vasospasm. Stroke. 32:2913–2919. 2001. View Article : Google Scholar : PubMed/NCBI

76 

Salomone S, Yoshimura S, Reuter U, Foley M, Thomas SS, Moskowitz MA and Waeber C: S1P3 receptors mediate the potent constriction of cerebral arteries by sphingosine-1-phosphate. Eur J Pharmacol. 469:125–134. 2003. View Article : Google Scholar : PubMed/NCBI

77 

Yogi A, Callera GE, Aranha AB, Antunes TT, Graham D, McBride M, Dominiczak A and Touyz RM: Sphingosine-1-phosphate-induced inflammation involves receptor tyrosine kinase transactivation in vascular cells: Upregulation in hypertension. Hypertension. 57:809–818. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2016
Volume 38 Issue 4

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen W, Lu H, Yang J, Xiang H and Peng H: Sphingosine 1-phosphate in metabolic syndrome (Review). Int J Mol Med 38: 1030-1038, 2016
APA
Chen, W., Lu, H., Yang, J., Xiang, H., & Peng, H. (2016). Sphingosine 1-phosphate in metabolic syndrome (Review). International Journal of Molecular Medicine, 38, 1030-1038. https://doi.org/10.3892/ijmm.2016.2731
MLA
Chen, W., Lu, H., Yang, J., Xiang, H., Peng, H."Sphingosine 1-phosphate in metabolic syndrome (Review)". International Journal of Molecular Medicine 38.4 (2016): 1030-1038.
Chicago
Chen, W., Lu, H., Yang, J., Xiang, H., Peng, H."Sphingosine 1-phosphate in metabolic syndrome (Review)". International Journal of Molecular Medicine 38, no. 4 (2016): 1030-1038. https://doi.org/10.3892/ijmm.2016.2731