1
|
Liu J, Liu X, Luo Y, Huang F, Xie Y, Zheng S, Jia B, Xiao Z. Sphingolipids: drivers of cardiac fibrosis and atrial fibrillation. J Mol Med (Berl) 2024; 102:149-165. [PMID: 38015241 PMCID: PMC10858135 DOI: 10.1007/s00109-023-02391-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 10/13/2023] [Accepted: 10/23/2023] [Indexed: 11/29/2023]
Abstract
Sphingolipids (SLs) are vital constituents of the plasma membrane of animal cells and concurrently regulate numerous cellular processes. An escalating number of research have evinced that SLs assume a crucial part in the progression of tissue fibrosis, a condition for which no efficacious cure exists as of now. Cardiac fibrosis, and in particular, atrial fibrosis, is a key factor in the emergence of atrial fibrillation (AF). AF has become one of the most widespread cardiac arrhythmias globally, with its incidence continuing to mount, thereby propelling it to the status of a major public health concern. This review expounds on the structure and biosynthesis pathways of several pivotal SLs, the pathophysiological mechanisms of AF, and the function of SLs in cardiac fibrosis. Delving into the influence of sphingolipid levels in the alleviation of cardiac fibrosis offers innovative therapeutic strategies to address cardiac fibrosis and AF.
Collapse
Affiliation(s)
- Junjie Liu
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ximao Liu
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yucheng Luo
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fangze Huang
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yu Xie
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shaoyi Zheng
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Bo Jia
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China.
| | - Zezhou Xiao
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
2
|
Kajita K, Ishii I, Mori I, Asano M, Fuwa M, Morita H. Sphingosine 1-Phosphate Regulates Obesity and Glucose Homeostasis. Int J Mol Sci 2024; 25:932. [PMID: 38256005 PMCID: PMC10816022 DOI: 10.3390/ijms25020932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/04/2024] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
One of the major global health and welfare issues is the treatment of obesity and associated metabolic disorders, such as type 2 diabetes mellitus and nonalcoholic fatty liver disease. Obesity, caused by the excessive accumulation of triglycerides in adipose tissues, induces adipocyte dysfunction, followed by inflammation, in adipose tissues and lipotoxicity in nonadipose tissues. Several studies have shown that obesity and glucose homeostasis are influenced by sphingolipid mediators, including ceramide and sphingosine 1-phosphate (S1P). Cellular accumulation of ceramide impairs pancreatic β-cell survival, confers insulin resistance in the liver and the skeletal muscle, and deteriorates adipose tissue inflammation via unknown molecular mechanisms. The roles of S1P are more complicated, because there are five cell-surface S1P receptors (S1PRs: S1P1-5) which have altered functions, different cellular expression patterns, and inapparent intracellular targets. Recent findings, including those by our group, support the notable concept that the pharmacological activation of S1P1 or S1P3 improves obesity and associated metabolic disorders, whereas that of S1P2 has the opposite effect. In addition, the regulation of S1P production by sphingosine kinase (SphK) is an essential factor affecting glucose homeostasis. This review summarizes the current knowledge on SphK/S1P/S1PR signaling in and against obesity, insulin resistance, and associated disorders.
Collapse
Affiliation(s)
- Kazuo Kajita
- Department of Health and Nutrition, Faculty of Home Economics, Gifu Women’s University, 80 Taromaru, Gifu 501-2592, Japan
| | - Isao Ishii
- Department of Health Chemistry, Showa Pharmaceutical University, 3-3165 Higashitamagawagakuen, Machida 194-8543, Japan
| | - Ichiro Mori
- Department of General Medicine and General Internal Medicine, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu 501-1194, Japan; (I.M.); (M.A.); (M.F.); (H.M.)
| | - Motochika Asano
- Department of General Medicine and General Internal Medicine, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu 501-1194, Japan; (I.M.); (M.A.); (M.F.); (H.M.)
| | - Masayuki Fuwa
- Department of General Medicine and General Internal Medicine, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu 501-1194, Japan; (I.M.); (M.A.); (M.F.); (H.M.)
| | - Hiroyuki Morita
- Department of General Medicine and General Internal Medicine, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu 501-1194, Japan; (I.M.); (M.A.); (M.F.); (H.M.)
| |
Collapse
|
3
|
Jiang H, Huang T, Yu Y, Zhou C, Qiu L, Mai HN, Gropler RJ, Klein RS, Tu Z. Characterization of a S1PR2 specific 11C-labeled radiotracer in streptozotocin-induced diabetic murine model. Nucl Med Biol 2023; 122-123:108370. [PMID: 37556928 PMCID: PMC10949307 DOI: 10.1016/j.nucmedbio.2023.108370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/21/2023] [Accepted: 07/24/2023] [Indexed: 08/11/2023]
Abstract
BACKGROUND Diabetes mellitus is a chronic progressive metabolic disorder that affects millions of people worldwide. Emerging evidence suggests the important roles of sphingolipid metabolism in diabetes. In particular, sphingosine-1-phosphate (S1P) and S1P receptor 2 (S1PR2) have important metabolic functions and are involved in several metabolic diseases. In diabetes, S1PR2 can effectively preserve β cells and improve glucose/insulin tolerance in high-fat diet induced and streptozotocin (STZ)-induced diabetic mouse models. We previously developed a group of potent and selective S1PR2 ligands and radioligands. METHODS In this study, we continued our efforts and characterized our leading S1PR2 radioligand, [11C]TZ34125, in a STZ-induced diabetic mouse model. [11C]TZ34125 was radiosynthesized in an automated synthesis module and in vitro saturation binding assay was performed using recombinant human S1PR2 membrane. In vitro saturation autoradiography analysis was also performed to determine the binding affinity of [11C]TZ34125 against mouse tissues. Type-1 diabetic mouse model was developed following a single high dose of STZ in C57BL/6 mice. Ex vivo biodistribution was performed to evaluate the distribution and amount of [11C]TZ34125 in tissues. In vitro autoradiography analysis was performed to compare the uptake of [11C]TZ34125 between diabetic and control animals in mouse spleen and pancreas. RESULTS Our in vitro saturation binding assay using [11C]TZ34125 confirmed [11C]TZ34125 is a potent radioligand to recombinant human S1PR2 membrane with a Kd value of 0.9 nM. Saturation autoradiographic analysis showed [11C]TZ34125 has a Kd of 67.5, 45.9, and 25.0 nM to mouse kidney, spleen, and liver tissues respectively. Biodistribution study in STZ-induced diabetic mice showed the uptake of [11C]TZ34125 was significantly elevated in the spleen (~2 fold higher) and pancreas (~1.4 fold higher) compared to normal controls. The increased uptake of [11C]TZ34125 was further confirmed using autoradiographic analysis in the spleen and pancreases of STZ-induced diabetic mice, indicating S1PR2 can potentially act as a biomarker of diabetes in pancreases and inflammation in spleen. Future mechanistic analysis and in vivo quantitative assessment using non-invasive PET imaging in large animal model of diabetes is worthwhile. CONCLUSIONS Overall, our data showed an increased uptake of our lead S1PR2-specific radioligand, [11C]TZ34125, in the spleen and pancreases of STZ-induced diabetic mice, and demonstrated [11C]TZ34125 has a great potential for preclinical and clinical usage for assessment of S1PR2 in diabetes and inflammation.
Collapse
Affiliation(s)
- Hao Jiang
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, 63110, United States of America
| | - Tianyu Huang
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, 63110, United States of America
| | - Yanbo Yu
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, 63110, United States of America
| | - Charles Zhou
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, 63110, United States of America
| | - Lin Qiu
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, 63110, United States of America
| | - Hien Ngoc Mai
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, 63110, United States of America
| | - Robert J Gropler
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, 63110, United States of America
| | - Robyn S Klein
- Departments of Medicine and Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, 63110, United States of America
| | - Zhude Tu
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, 63110, United States of America.
| |
Collapse
|
4
|
Duan M, Gao P, Chen SX, Novák P, Yin K, Zhu X. Sphingosine-1-phosphate in mitochondrial function and metabolic diseases. Obes Rev 2022; 23:e13426. [PMID: 35122459 DOI: 10.1111/obr.13426] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/02/2022] [Accepted: 01/02/2022] [Indexed: 01/23/2023]
Abstract
Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid metabolite. The past decade has witnessed exponential growth in the field of S1P research, partly attributed to drugs targeting its receptors or kinases. Accumulating evidence indicates that changes in the S1P axis (i.e., S1P production, transport, and receptors) may modify metabolism and eventually mediate metabolic diseases. Dysfunction of the mitochondria on a master monitor of cellular metabolism is considered the leading cause of metabolic diseases, with aberrations typically induced by abnormal biogenesis, respiratory chain complex disorders, reactive oxygen species overproduction, calcium deposition, and mitophagy impairment. Accordingly, we discuss decades of investigation into changes in the S1P axis and how it controls mitochondrial function. Furthermore, we summarize recent scientific advances in disorders associated with the S1P axis and their involvement in the pathogenesis of metabolic diseases in humans, including type 2 diabetes mellitus and cardiovascular disease, from the perspective of mitochondrial function. Finally, we review potential challenges and prospects for S1P axis application to the regulation of mitochondrial function and metabolic diseases; these data may provide theoretical guidance for the treatment of metabolic diseases.
Collapse
Affiliation(s)
- Meng Duan
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi, China
| | - Pan Gao
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi, China
| | - Sheng-Xi Chen
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi, China
| | - Petr Novák
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi, China
| | - Kai Yin
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi, China.,Department of Cardiology, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Xiao Zhu
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi, China
| |
Collapse
|
5
|
Luo Z, Liu H, Yu Y, Gropler RJ, Klein RS, Tu Z. Synthesis and evaluation of highly selective quinazoline-2,4-dione ligands for sphingosine-1-phosphate receptor 2. RSC Med Chem 2022; 13:202-207. [PMID: 35308025 PMCID: PMC8864552 DOI: 10.1039/d1md00357g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 12/22/2021] [Indexed: 01/05/2023] Open
Abstract
A series of twenty-nine new quinazoline-2,4-dione compounds were synthesized and their IC50 values for binding toward sphingosine-1-phosphate receptor 2 (S1PR2) were determined using a [32P]S1P binding assay. Seven compounds 2a, 2g, 2h, 2i, 2j, 2k, and 5h exhibit high S1PR2 binding potencies (IC50 values < 50 nM) and four of these new compounds 2g, 2i, 2j, and 2k have IC50 values (<10 nM) of 6.3, 5.7, 4.8, and 2.6 nM, and are highly selective for S1PR2 over other S1PR subtypes, S1PR1, 3, 4, and 5. Compounds 2a and 2i were chosen for C-11 radiosynthesis through O-[11C]methylation of precursors 13 and 2k with good radiochemical yields (35-40%), high chemical and radiochemical purity (>98%), and high molar activity (153-222 GBq μmol-1, at the end of bombardment). [11C]2a and [11C]2i were further evaluated by the ex vivo biodistribution study. The results showed that both tracers have low brain uptake, preventing their potential for neuroimaging application. Further explorations of this class of S1PR2 PET tracers in peripheral tissue diseases are underway.
Collapse
Affiliation(s)
- Zonghua Luo
- Department of Radiology, Washington University School of Medicine St. Louis MO 63110 USA
- School of Biomedical Engineering, ShanghaiTech University Shanghai 201210 China
| | - Hui Liu
- Department of Radiology, Washington University School of Medicine St. Louis MO 63110 USA
| | - Yanbo Yu
- Department of Radiology, Washington University School of Medicine St. Louis MO 63110 USA
| | - Robert J Gropler
- Department of Radiology, Washington University School of Medicine St. Louis MO 63110 USA
| | - Robyn S Klein
- Department of Medicine, Washington University School of Medicine St. Louis MO 63110 USA
- Department of Neuroscience, Washington University School of Medicine St. Louis MO 63110 USA
- Department of Pathology & Immunology, Washington University School of Medicine St. Louis MO 63110 USA
| | - Zhude Tu
- Department of Radiology, Washington University School of Medicine St. Louis MO 63110 USA
| |
Collapse
|
6
|
Gurgul-Convey E. To Be or Not to Be: The Divergent Action and Metabolism of Sphingosine-1 Phosphate in Pancreatic Beta-Cells in Response to Cytokines and Fatty Acids. Int J Mol Sci 2022; 23:ijms23031638. [PMID: 35163559 PMCID: PMC8835924 DOI: 10.3390/ijms23031638] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/24/2022] [Accepted: 01/27/2022] [Indexed: 01/02/2023] Open
Abstract
Sphingosine-1 phosphate (S1P) is a bioactive sphingolipid with multiple functions conveyed by the activation of cell surface receptors and/or intracellular mediators. A growing body of evidence indicates its important role in pancreatic insulin-secreting beta-cells that are necessary for maintenance of glucose homeostasis. The dysfunction and/or death of beta-cells lead to diabetes development. Diabetes is a serious public health burden with incidence growing rapidly in recent decades. The two major types of diabetes are the autoimmune-mediated type 1 diabetes (T1DM) and the metabolic stress-related type 2 diabetes (T2DM). Despite many differences in the development, both types of diabetes are characterized by chronic hyperglycemia and inflammation. The inflammatory component of diabetes remains under-characterized. Recent years have brought new insights into the possible mechanism involved in the increased inflammatory response, suggesting that environmental factors such as a westernized diet may participate in this process. Dietary lipids, particularly palmitate, are substrates for the biosynthesis of bioactive sphingolipids. Disturbed serum sphingolipid profiles were observed in both T1DM and T2DM patients. Many polymorphisms were identified in genes encoding enzymes of the sphingolipid pathway, including sphingosine kinase 2 (SK2), the S1P generating enzyme which is highly expressed in beta-cells. Proinflammatory cytokines and free fatty acids have been shown to modulate the expression and activity of S1P-generating and S1P-catabolizing enzymes. In this review, the similarities and differences in the action of extracellular and intracellular S1P in beta-cells exposed to cytokines or free fatty acids will be identified and the outlook for future research will be discussed.
Collapse
Affiliation(s)
- Ewa Gurgul-Convey
- Institute of Clinical Biochemistry, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| |
Collapse
|
7
|
Thomas JM, Sudhadevi T, Basa P, Ha AW, Natarajan V, Harijith A. The Role of Sphingolipid Signaling in Oxidative Lung Injury and Pathogenesis of Bronchopulmonary Dysplasia. Int J Mol Sci 2022; 23:ijms23031254. [PMID: 35163176 PMCID: PMC8835774 DOI: 10.3390/ijms23031254] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 02/07/2023] Open
Abstract
Premature infants are born with developing lungs burdened by surfactant deficiency and a dearth of antioxidant defense systems. Survival rate of such infants has significantly improved due to advances in care involving mechanical ventilation and oxygen supplementation. However, a significant subset of such survivors develops the chronic lung disease, Bronchopulmonary dysplasia (BPD), characterized by enlarged, simplified alveoli and deformed airways. Among a host of factors contributing to the pathogenesis is oxidative damage induced by exposure of the developing lungs to hyperoxia. Recent data indicate that hyperoxia induces aberrant sphingolipid signaling, leading to mitochondrial dysfunction and abnormal reactive oxygen species (ROS) formation (ROS). The role of sphingolipids such as ceramides and sphingosine 1-phosphate (S1P), in the development of BPD emerged in the last decade. Both ceramide and S1P are elevated in tracheal aspirates of premature infants of <32 weeks gestational age developing BPD. This was faithfully reflected in the murine models of hyperoxia and BPD, where there is an increased expression of sphingolipid metabolites both in lung tissue and bronchoalveolar lavage. Treatment of neonatal pups with a sphingosine kinase1 specific inhibitor, PF543, resulted in protection against BPD as neonates, accompanied by improved lung function and reduced airway remodeling as adults. This was accompanied by reduced mitochondrial ROS formation. S1P receptor1 induced by hyperoxia also aggravates BPD, revealing another potential druggable target in this pathway for BPD. In this review we aim to provide a detailed description on the role played by sphingolipid signaling in hyperoxia induced lung injury and BPD.
Collapse
Affiliation(s)
- Jaya M. Thomas
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, USA; (J.M.T.); (T.S.); (P.B.); (A.W.H.)
| | - Tara Sudhadevi
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, USA; (J.M.T.); (T.S.); (P.B.); (A.W.H.)
| | - Prathima Basa
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, USA; (J.M.T.); (T.S.); (P.B.); (A.W.H.)
| | - Alison W. Ha
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, USA; (J.M.T.); (T.S.); (P.B.); (A.W.H.)
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Viswanathan Natarajan
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA;
- Department of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Anantha Harijith
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, USA; (J.M.T.); (T.S.); (P.B.); (A.W.H.)
- Correspondence: ; Tel.: +1-(216)-286-7038
| |
Collapse
|
8
|
Lin S, Pandruvada S, Yu H. Inhibition of Sphingosine-1-Phosphate Receptor 2 by JTE013 Promoted Osteogenesis by Increasing Vesicle Trafficking, Wnt/Ca 2+, and BMP/Smad Signaling. Int J Mol Sci 2021; 22:ijms222112060. [PMID: 34769490 PMCID: PMC8584480 DOI: 10.3390/ijms222112060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 10/25/2021] [Accepted: 11/05/2021] [Indexed: 12/31/2022] Open
Abstract
Sphingosine-1-phosphate receptor 2 (S1PR2) is a G protein-coupled receptor that regulates various immune responses. Herein, we determine the effects of a S1PR2 antagonist (JTE013) or a S1PR2 shRNA on osteogenesis by culturing murine bone marrow stromal cells (BMSCs) in osteogenic media with JTE013, dimethylsulfoxide (DMSO), a S1PR2 shRNA, or a control shRNA. Treatment with JTE013 or the S1PR2 shRNA increased alkaline phosphatase and alizarin red s staining, and enhanced alkaline phosphatase, RUNX2, osteocalcin, and osterix mRNA levels in BMSCs compared with the controls. Protein analysis revealed that a high dose of JTE013 (4 or 8 μM) increased vesicle trafficking-associated proteins (F-actin, clathrin, Early Endosome Antigen 1 (EEA1), and syntaxin 6) and Wnt/Ca2+ signaling. On the other hand, a low dose of JTE013 (1 to 2 μM) increased BMP/Smad signaling. In contrast, the S1PR2 shRNA reduced vesicle trafficking-associated proteins and attenuated Wnts and BMP/Smad signaling, but enhanced p-CaMKII compared with the control, suggesting that the S1PR2 shRNA influenced osteogenesis via different signaling pathways. Moreover, inhibiting protein trafficking by brefeldin A in BMSCs suppressed Wnts and BMPRs expressions. These data supported that enhanced osteogenesis in JTE013-treated BMSCs is associated with increased vesicle trafficking, which promotes the synthesis and transport of osteogenic protein and matrix vesicles and enhances matrix mineralization.
Collapse
|
9
|
Rosa LRDO, Vettorazzi JF, Zangerolamo L, Carneiro EM, Barbosa HCDL. TUDCA receptors and their role on pancreatic beta cells. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2021; 167:26-31. [PMID: 34547326 DOI: 10.1016/j.pbiomolbio.2021.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 08/31/2021] [Accepted: 09/14/2021] [Indexed: 12/12/2022]
Abstract
Bile acids have received increasing attention over the past years as their multiple alternative roles became clearer. Tauroursodeoxycholic Acid (TUDCA) in specific has generated special interest due to its ability to promote pancreatic survival and function, as well as reduce endoplasmic reticulum stress. However, there are few studies explaining the molecular mechanisms behind TUDCA's beneficial actions on pancreatic beta cells. In this review, we decided to review the literature in order to craft a primer for researchers on what is known about TUDCA's receptors and the molecular pathways involved in this bile acid's function in the endocrine pancreas. We review the studies that focused on G protein-coupled bile acid receptor (TGR5), Sphingosine-1-phosphate receptor 2 (S1PR2) and α5β1 Integrin function in pancreatic cells. Our hope is to provide a basis for future studies to expand upon, especially considering the current lack of studies focusing on the importance of these receptors, either through TUDCA signaling or other signaling molecules.
Collapse
Affiliation(s)
- Lucas Rodolfo de Oliveira Rosa
- Obesity and Comorbidities Research Center (OCRC), Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.
| | | | - Lucas Zangerolamo
- Obesity and Comorbidities Research Center (OCRC), Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.
| | - Everardo Magalhães Carneiro
- Obesity and Comorbidities Research Center (OCRC), Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.
| | - Helena Cristina de Lima Barbosa
- Obesity and Comorbidities Research Center (OCRC), Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.
| |
Collapse
|
10
|
Wigger D, Schumacher F, Schneider-Schaulies S, Kleuser B. Sphingosine 1-phosphate metabolism and insulin signaling. Cell Signal 2021; 82:109959. [PMID: 33631318 DOI: 10.1016/j.cellsig.2021.109959] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/17/2021] [Accepted: 02/18/2021] [Indexed: 12/19/2022]
Abstract
Insulin is the main anabolic hormone secreted by β-cells of the pancreas stimulating the assimilation and storage of glucose in muscle and fat cells. It modulates the postprandial balance of carbohydrates, lipids and proteins via enhancing lipogenesis, glycogen and protein synthesis and suppressing glucose generation and its release from the liver. Resistance to insulin is a severe metabolic disorder related to a diminished response of peripheral tissues to the insulin action and signaling. This leads to a disturbed glucose homeostasis that precedes the onset of type 2 diabetes (T2D), a disease reaching epidemic proportions. A large number of studies reported an association between elevated circulating fatty acids and the development of insulin resistance. The increased fatty acid lipid flux results in the accumulation of lipid droplets in a variety of tissues. However, lipid intermediates such as diacylglycerols and ceramides are also formed in response to elevated fatty acid levels. These bioactive lipids have been associated with the pathogenesis of insulin resistance. More recently, sphingosine 1-phosphate (S1P), another bioactive sphingolipid derivative, has also been shown to increase in T2D and obesity. Although many studies propose a protective role of S1P metabolism on insulin signaling in peripheral tissues, other studies suggest a causal role of S1P on insulin resistance. In this review, we critically summarize the current state of knowledge of S1P metabolism and its modulating role on insulin resistance. A particular emphasis is placed on S1P and insulin signaling in hepatocytes, skeletal muscle cells, adipocytes and pancreatic β-cells. In particular, modulation of receptors and enzymes that regulate S1P metabolism can be considered as a new therapeutic option for the treatment of insulin resistance and T2D.
Collapse
Affiliation(s)
- Dominik Wigger
- Institute of Pharmacy, Pharmacology and Toxicology, Freie Universität Berlin, Berlin, Germany; Institute of Nutritional Science, Nutritional Toxicology, University of Potsdam, Nuthetal, Germany
| | - Fabian Schumacher
- Institute of Pharmacy, Pharmacology and Toxicology, Freie Universität Berlin, Berlin, Germany; Institute of Nutritional Science, Nutritional Toxicology, University of Potsdam, Nuthetal, Germany
| | | | - Burkhard Kleuser
- Institute of Pharmacy, Pharmacology and Toxicology, Freie Universität Berlin, Berlin, Germany; Institute of Nutritional Science, Nutritional Toxicology, University of Potsdam, Nuthetal, Germany.
| |
Collapse
|
11
|
Luo Z, Liang Q, Liu H, Sumit J, Jiang H, Klein RS, Tu Z. Synthesis and characterization of [ 125I]TZ6544, a promising radioligand for investigating sphingosine-1-phosphate receptor 2. Nucl Med Biol 2020; 88-89:52-61. [PMID: 32791475 DOI: 10.1016/j.nucmedbio.2020.07.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 06/14/2020] [Accepted: 07/26/2020] [Indexed: 01/20/2023]
Abstract
INTRODUCTION Sphingosine-1-phosphate receptor 2 (S1PR2) activation exerts a critical role in biological abnormalities and diseases. A suitable radiotracer will advance our understanding of S1PR2 pathophysiology of diseases. The objective of this study is to evaluate the potential of iodine-125 labeled [125I]TZ6544 to be used for screening new compounds binding toward S1PR2, and assessing the changes of S1PR2 expression in the kidney of streptozotocin-induced diabetic rats. METHODS [125I]TZ6544 was synthesized from borate precursor by copper (II)-catalyzed iodization reaction with [125I]NaI. [125I]TZ6544 was characterized using human recombinant S1PR2 cell membrane and biodistribution studies of [125]TZ6544 were performed on Wistar rats that were euthanized at 5 and 30 min post-injection. A rat model of diabetes was induced by IV injection of streptozotocin (55 mg/kg). In vitro autoradiography studies, immunostaining, and enzyme-linked immunosorbent assay (ELISA) analysis were performed in both diabetic and control rats. RESULTS Radiosynthesis of [125I]TZ6544 was achieved successfully with good radiochemical yields of ~47% and high radiochemical purity of >99%. [125I]TZ6544 is a potent ligand in vitro for S1PR2 with Kd value of 4.31 nM. [125I]TZ6544 and [32P]-labeled endogenous S1P provided comparable IC50 values in radioactive competitive binding assays against known S1PR2 ligands. Compared to control, the kidney of diabetic rats had increased uptake of [125I]TZ6544, which could be reduced by a S1PR2 antagonist, JTE-013. Immunostaining and ELISA analysis confirmed that the diabetic rat had increased S1PR2 expression in the kidney. CONCLUSIONS [125I]TZ6544 was synthesized successfully in high yields, and in vitro evaluation suggested [125I]TZ6544 has high potential to be used for screening new S1PR2 compounds and investigating the pathophysiology of S1PR2 functions. The availability of [125I]TZ6544 may facilitate the development of therapeutics and imaging agents targeting S1PR2. ADVANCES IN KNOWLEDGE: [125I]TZ6544 showed increased expression of S1PR2 in diabetic rat kidney and can be used to determine binding potency of S1PR2 compounds.
Collapse
Affiliation(s)
- Zonghua Luo
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Qianwa Liang
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hui Liu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Joshi Sumit
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hao Jiang
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Robyn S Klein
- Departments of Medicine, Neuroscience, Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Zhude Tu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
12
|
Kim SJ, Miller B, Mehta HH, Xiao J, Wan J, Arpawong TE, Yen K, Cohen P. The mitochondrial-derived peptide MOTS-c is a regulator of plasma metabolites and enhances insulin sensitivity. Physiol Rep 2020; 7:e14171. [PMID: 31293078 PMCID: PMC6640593 DOI: 10.14814/phy2.14171] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 06/19/2019] [Accepted: 06/20/2019] [Indexed: 12/16/2022] Open
Abstract
MOTS‐c is an exercise mimetic and improves insulin sensitivity in aged and diet‐induced obese mice. Although plasma markers are good markers for the metabolic condition, whether MOTS‐c changes plasma markers in diet‐induced obese mice has not been examined. Here, we used an unbiased metabolomics approach to examine the effect of MOTS‐c on plasma markers of metabolic dysfunction. We found that three pathways – sphingolipid metabolism, monoacylglycerol metabolism, and dicarboxylate metabolism – were reduced in MOTS‐c–injected mice. Interestingly, these pathways are upregulated in obese and T2D models. MOTS‐c improves insulin sensitivity and increases beta‐oxidation to prevent fat accumulation in DIO mice through these pathways. These results provide us a better understanding of the mechanism of how MOTS‐c improves insulin sensitivity and reduces the body weight and fatty liver and opens a new venue for further study.
Collapse
Affiliation(s)
- Su-Jeong Kim
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA
| | - Brendan Miller
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA
| | - Hemal H Mehta
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA
| | - Jialin Xiao
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA
| | - Junxiang Wan
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA
| | - Thalida E Arpawong
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA
| | - Kelvin Yen
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA
| | - Pinchas Cohen
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA
| |
Collapse
|
13
|
Funcke JB, Scherer PE. Beyond adiponectin and leptin: adipose tissue-derived mediators of inter-organ communication. J Lipid Res 2019; 60:1648-1684. [PMID: 31209153 PMCID: PMC6795086 DOI: 10.1194/jlr.r094060] [Citation(s) in RCA: 187] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 06/17/2019] [Indexed: 01/10/2023] Open
Abstract
The breakthrough discoveries of leptin and adiponectin more than two decades ago led to a widespread recognition of adipose tissue as an endocrine organ. Many more adipose tissue-secreted signaling mediators (adipokines) have been identified since then, and much has been learned about how adipose tissue communicates with other organs of the body to maintain systemic homeostasis. Beyond proteins, additional factors, such as lipids, metabolites, noncoding RNAs, and extracellular vesicles (EVs), released by adipose tissue participate in this process. Here, we review the diverse signaling mediators and mechanisms adipose tissue utilizes to relay information to other organs. We discuss recently identified adipokines (proteins, lipids, and metabolites) and briefly outline the contributions of noncoding RNAs and EVs to the ever-increasing complexities of adipose tissue inter-organ communication. We conclude by reflecting on central aspects of adipokine biology, namely, the contribution of distinct adipose tissue depots and cell types to adipokine secretion, the phenomenon of adipokine resistance, and the capacity of adipose tissue to act both as a source and sink of signaling mediators.
Collapse
Affiliation(s)
- Jan-Bernd Funcke
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX
| | - Philipp E Scherer
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
14
|
He Y, Shi B, Zhao X, Sui J. Sphingosine-1-phosphate induces islet β-cell proliferation and decreases cell apoptosis in high-fat diet/streptozotocin diabetic mice. Exp Ther Med 2019; 18:3415-3424. [PMID: 31602216 PMCID: PMC6777293 DOI: 10.3892/etm.2019.7999] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Accepted: 08/08/2019] [Indexed: 12/17/2022] Open
Abstract
Sphingosine-1-phosphate (S1P) has been reported to enhance the function of islet β-cells, providing a potential therapeutic target for diabetes mellitus. In the present study, the effects of S1P on the proliferation and apoptosis of β-cells in type 2 diabetic mice were investigated. The mice were administered intraperitoneal S1P solution daily at a dose of 20 µg/kg for three weeks. The intraperitoneal glucose tolerance test (IPGTT) and homeostatic model assessment of insulin resistance (HOMA-IR) index determination were carried out. Immunohistochemical staining was used to detect the protein expression of insulin, antigen Ki-67 and S1P receptor isoforms (S1PR1/S1PR2/S1PR3) in pancreatic islets. Compared with the diabetic control (DC) group, the IPGTT results and HOMA-IR index in the S1P treatment group were decreased. The islets in the S1P group exhibited higher insulin immunostaining intensity than the DC group, as well as higher proliferation (P<0.05) and lower apoptosis rates (P<0.05). Positive staining for the S1P receptors S1PR1, S1PR2 and S1PR3 was observed in the cytoplasm and membrane of the islet cells. S1PR1 and S1PR2 proteins showed increased expression in the S1P and DC groups compared with the normal control group (P<0.01 and P<0.05, respectively), whereas no significant difference was observed in the expression of S1PR3 among these groups. In conclusion, extracellular S1P can induce islet β-cell proliferation and decrease cell apoptosis in diabetic mice. S1P function may be mediated via S1PR1 and S1PR2; therefore, targeting S1P/S1PR signalling pathways may be a novel therapeutic strategy for diabetes mellitus.
Collapse
Affiliation(s)
- Yizhi He
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China.,Department of Endocrinology, Xi'an No. 3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, Shaanxi 710018, P.R. China
| | - Bingyin Shi
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xinrui Zhao
- Department of Immunology and Rheumatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Jing Sui
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
15
|
Design, synthesis, and in vitro bioactivity evaluation of fluorine-containing analogues for sphingosine-1-phosphate 2 receptor. Bioorg Med Chem 2019; 27:3619-3631. [PMID: 31279524 PMCID: PMC6698139 DOI: 10.1016/j.bmc.2019.06.047] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 06/18/2019] [Accepted: 06/28/2019] [Indexed: 12/15/2022]
Abstract
Twenty eight new aryloxybenzene analogues were synthesized and their in vitro binding potencies toward S1PR2 were determined using a [32P]S1P competitive binding assay. Out of these new analogues, three compounds, 28c (IC50 = 29.9 ± 3.9 nM), 28e (IC50 = 14.6 ± 1.5 nM), and 28g (IC50 = 38.5 ± 6.3 nM) exhibited high binding potency toward S1PR2 and high selectivity over the other four receptor subtypes (S1PR1, 3, 4, and 5; IC50 > 1000 nM). Each of the three potent compounds 28c, 28e, and 28g contains a fluorine atom that will allow to develop F-18 labeled PET radiotracers for imaging S1PR2.
Collapse
|
16
|
Nejatian N, Trautmann S, Thomas D, Pfeilschifter J, Badenhoop K, Koch A, Penna-Martinez M. Vitamin D effects on sphingosine 1-phosphate signaling and metabolism in monocytes from type 2 diabetes patients and controls. J Steroid Biochem Mol Biol 2019; 186:130-135. [PMID: 30336275 DOI: 10.1016/j.jsbmb.2018.10.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 09/17/2018] [Accepted: 10/06/2018] [Indexed: 12/21/2022]
Abstract
Elevated sphingosine 1-phopshate (S1P) concentration was observed in type 2 diabetes mellitus (T2D). On the other side, 1α,25-dihydroxyvitamin D3 (1,25(OH)2D3) can influence the formation of sphingosine 1-phopshate (S1P) and the expression of S1P receptors, which are known to be involved in T2D. In order to evaluate mechanisms for the antiinflammatory potential of 1,25(OH)2D3, we investigated whether 1,25(OH)2D3 alters S1P signaling and metabolism in human CD14+ monocytes. Primary monocytes isolated from healthy controls (HC) and T2D patients were treated for 24 h with 10 nM 1,25(OH)2D3 in the absence or presence of 500 IU/ml interleukin-(IL)-1β. Thereafter, sphingosine kinase (SPHK)1, SPHK2 and S1P receptor 1-5 (S1P1-5) mRNA expression levels were measured by TaqMan™ analyses. Sphingolipid levels in cell supernatant were determined by high-performance liquid chromatography/tandem mass spectrometry (LC-MS/MS). 1,25(OH)2D3 treatment downregulated S1P1 and S1P2 mRNA expression compared to untreated monocytes of HC and T2D patients. In contrast, SPHK1, S1P3 and S1P4 mRNA expression levels were upregulated by 1,25(OH)2D3 treatment compared to the respective controls. Furthermore, reduced S1P2 and increased S1P3 and S1P4 mRNA expression levels upon treatment with 1,25(OH)2D3 occurred in the presence of IL-1β. Additionally, S1P levels in cell supernatants were decreased in monocytes from HC and T2D patients by 1,25(OH)2D3 with or without IL-1β costimulation. The levels of sphingosine in cell supernatants were not influenced by 1,25(OH)2D3. Overall, our results demonstrate for the first time that 1,25(OH)2D3 treatment can influence S1P receptor and SPHK expression and S1P levels in primary monocytes of both HC and subjects with T2D. These findings justify further investigations into the sphingolipid metabolism and potential benefits of vitamin D treatment in diabetes.
Collapse
Affiliation(s)
- Nojan Nejatian
- Department of Internal Medicine I, Division of Endocrinology, Diabetes and Metabolism, Goethe University Hospital, Frankfurt am Main, Germany.
| | - Sandra Trautmann
- Department of Clinical Pharmacology, Goethe University Hospital, Frankfurt am Main, Germany
| | - Dominique Thomas
- Department of Clinical Pharmacology, Goethe University Hospital, Frankfurt am Main, Germany
| | - Josef Pfeilschifter
- Department of General Pharmacology and Toxicology, Goethe University Hospital, Frankfurt am Main, Germany
| | - Klaus Badenhoop
- Department of Internal Medicine I, Division of Endocrinology, Diabetes and Metabolism, Goethe University Hospital, Frankfurt am Main, Germany
| | - Alexander Koch
- Department of General Pharmacology and Toxicology, Goethe University Hospital, Frankfurt am Main, Germany
| | - Marissa Penna-Martinez
- Department of Internal Medicine I, Division of Endocrinology, Diabetes and Metabolism, Goethe University Hospital, Frankfurt am Main, Germany
| |
Collapse
|
17
|
Chu C, Li D, Zhang S, Ikejima T, Jia Y, Wang D, Xu F. Role of silibinin in the management of diabetes mellitus and its complications. Arch Pharm Res 2018; 41:785-796. [DOI: 10.1007/s12272-018-1047-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Accepted: 06/16/2018] [Indexed: 02/07/2023]
|
18
|
Bowers DT, Olingy CE, Chhabra P, Langman L, Merrill PH, Linhart RS, Tanes ML, Lin D, Brayman KL, Botchwey EA. An engineered macroencapsulation membrane releasing FTY720 to precondition pancreatic islet transplantation. J Biomed Mater Res B Appl Biomater 2018; 106:555-568. [PMID: 28240814 PMCID: PMC5572559 DOI: 10.1002/jbm.b.33862] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 12/28/2016] [Accepted: 01/26/2017] [Indexed: 02/06/2023]
Abstract
Macroencapsulation is a powerful approach to increase the efficiency of extrahepatic pancreatic islet transplant. FTY720, a small molecule that activates signaling through sphingosine-1-phosphate receptors, is immunomodulatory and pro-angiogenic upon sustained delivery from biomaterials. While FTY720 (fingolimod, Gilenya) has been explored for organ transplantation, in the present work the effect of locally released FTY720 from novel nanofiber-based macroencapsulation membranes is explored for islet transplantation. We screened islet viability during culture with FTY720 and various biodegradable polymers. Islet viability is significantly reduced by the addition of high doses (≥500 ng/mL) of soluble FTY720. Among the polymers screened, islets have the highest viability when cultured with poly(3-hydroxybutyrate-co-3-hydroxyvalerate) (PHBV). Therefore, PHBV was blended with polycaprolactone (PCL) for mechanical stability and electrospun into nanofibers. Islets had no detectable function ex vivo following 5 days or 12 h of subcutaneous implantation within our engineered device. Subsequently, we explored a preconditioning scheme in which islets are transplanted 2 weeks after FTY720-loaded nanofibers are implanted. This allows FTY720 to orchestrate a local regenerative milieu while preventing premature transplantation into avascular sites that contain high concentrations of FTY720. These results provide a foundation and motivation for further investigation into the use of FTY720 in preconditioning sites for efficacious islet transplantation. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 106B: 555-568, 2018.
Collapse
Affiliation(s)
- Daniel T Bowers
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, 22903
| | - Claire E Olingy
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, 30332-0363
| | - Preeti Chhabra
- Department of Surgery, University of Virginia, Charlottesville, Virginia, 22903
| | - Linda Langman
- Department of Surgery, University of Virginia, Charlottesville, Virginia, 22903
| | - Parker H Merrill
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, 22903
| | - Ritu S Linhart
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, 22903
| | - Michael L Tanes
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, 22903
| | - Dan Lin
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, 22903
| | - Kenneth L Brayman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, 22903
- Department of Surgery, University of Virginia, Charlottesville, Virginia, 22903
| | - Edward A Botchwey
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, 22903
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, 30332-0363
| |
Collapse
|
19
|
Arish M, Alaidarous M, Ali R, Akhter Y, Rub A. Implication of sphingosine-1-phosphate signaling in diseases: molecular mechanism and therapeutic strategies. J Recept Signal Transduct Res 2017; 37:437-446. [PMID: 28758826 DOI: 10.1080/10799893.2017.1358282] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Sphingosine-1-phosphate signaling is emerging as a critical regulator of cellular processes that is initiated by the intracellular production of bioactive lipid molecule, sphingosine-1-phosphate. Binding of sphingosine-1-phosphate to its extracellular receptors activates diverse downstream signaling that play a critical role in governing physiological processes. Increasing evidence suggests that this signaling pathway often gets impaired during pathophysiological and diseased conditions and hence manipulation of this signaling pathway may be beneficial in providing treatment. In this review, we summarized the recent findings of S1P signaling pathway and the versatile role of the participating candidates in context with several disease conditions. Finally, we discussed its possible role as a novel drug target in different diseases.
Collapse
Affiliation(s)
- Mohd Arish
- a Infection and Immunity Lab, Department of Biotechnology , Jamia Millia Islamia (A Central University) , New Delhi , India
| | - Mohammed Alaidarous
- b Department of Medical Laboratory Sciences, College of Applied Medical Sciences , Majmaah University , Al Majmaah , Saudi Arabia
| | - Rahat Ali
- a Infection and Immunity Lab, Department of Biotechnology , Jamia Millia Islamia (A Central University) , New Delhi , India
| | - Yusuf Akhter
- c Centre for Computational Biology & Bioinformatics, School of Life Sciences , Central University of Himachal Pradesh , Shahpur, Kangra , India
| | - Abdur Rub
- a Infection and Immunity Lab, Department of Biotechnology , Jamia Millia Islamia (A Central University) , New Delhi , India.,b Department of Medical Laboratory Sciences, College of Applied Medical Sciences , Majmaah University , Al Majmaah , Saudi Arabia
| |
Collapse
|
20
|
S1PR2 antagonist protects endothelial cells against high glucose-induced mitochondrial apoptosis through the Akt/GSK-3β signaling pathway. Biochem Biophys Res Commun 2017; 490:1119-1124. [PMID: 28676392 DOI: 10.1016/j.bbrc.2017.06.189] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 06/30/2017] [Indexed: 11/23/2022]
Abstract
Vascular complications are the main cause of morbidity and mortality associated with type 2 diabetes mellitus. An early hallmark of the onset of vascular complications is endothelial dysfunction and apoptosis. We aimed to explore the role of sphingosine-1-phosphatereceptor 2 (S1PR2) in high glucose-induced endothelial cells apoptosis and to elaborate the underlying mechanism. Human umbilical vein endothelial cells (HUVECs) were cultured in a high glucose with or without S1PR2 antagonist. The apoptosis of the cells was measured by flow cytometry and mitochondrial membrane permeability was detected by the fluorescent probe JC-1. The expression of the related protein was determined by western blot. Cell apoptosis and the loss of mitochondrial membrane permeability were induced under high glucose conditions in HUVECs. The expression of mitochondrial apoptosis related protein bax increased and bcl-2 decreased in high glucose-induced HUVECs. The level of cytochrome c released into the cytoplasm increased when cells were exposed to high glucose. In addition, the expression of p-AKT and p-GSK3β was reduced when HUVECs were treated with high glucose. However, these effects were reversed in HUVECs when cells treated with S1PR2 antagonist. In conclusion, S1PR2 antagonist protects endothelial cells against high glucose-induced mitochondrial apoptosis through the Akt/GSK-3β signaling pathway.
Collapse
|
21
|
Ng ML, Wadham C, Sukocheva OA. The role of sphingolipid signalling in diabetes‑associated pathologies (Review). Int J Mol Med 2017; 39:243-252. [PMID: 28075451 PMCID: PMC5358714 DOI: 10.3892/ijmm.2017.2855] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 11/14/2016] [Indexed: 02/05/2023] Open
Abstract
Sphingosine kinase (SphK) is an important signalling enzyme that catalyses the phosphorylation of sphingosine (Sph) to form sphingosine‑1‑phosphate (S1P). The multifunctional lipid, S1P binds to a family of five G protein-coupled receptors (GPCRs). As an intracellular second messenger, S1P activates key signalling cascades responsible for the maintenance of sphingolipid metabolism, and has been implicated in the progression of cancer, and the development of other inflammatory and metabolic diseases. SphK and S1P are critical molecules involved in the regulation of various cellular metabolic processes, such as cell proliferation, survival, apoptosis, adhesion and migration. There is strong evidence supporting the critical roles of SphK and S1P in the progression of diabetes mellitus, including insulin sensitivity and insulin secretion, pancreatic β‑cell apoptosis, and the development of diabetic inflammatory state. In this review, we summarise the current state of knowledge for SphK/S1P signalling effects, associated with the development of insulin resistance, pancreatic β‑cell death and the vascular complications of diabetes mellitus.
Collapse
Affiliation(s)
- Mei Li Ng
- Centenary Institute of Cancer Medicine and Cell Biology, Sydney, NSW 2050
- Sydney Medical School, Faculty of Medicine, University of Sydney, Sydney, NSW 2006, Australia
- Advanced Medical and Dental Institute, University Sains Malaysia, Kepala Batas, Penang 13200, Malaysia
- Correspondence to: Dr Mei Li Ng, Advanced Medical and Dental Institute, University Sains Malaysia, No. 1-8 (Lot 8), Persiaran Seksyen 4, 1, Bandar Putra Bertam, Kepala Batas, Penang 13200, Malaysia, E-mail:
| | - Carol Wadham
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales, Randwick, NSW 2031
| | - Olga A. Sukocheva
- School of Social Health Sciences, Flinders University, Bedford Park, SA 5042, Australia
| |
Collapse
|
22
|
Park SJ, Im DS. Sphingosine 1-Phosphate Receptor Modulators and Drug Discovery. Biomol Ther (Seoul) 2017; 25:80-90. [PMID: 28035084 PMCID: PMC5207465 DOI: 10.4062/biomolther.2016.160] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 10/06/2016] [Accepted: 10/27/2016] [Indexed: 01/07/2023] Open
Abstract
Initial discovery on sphingosine 1-phosphate (S1P) as an intracellular second messenger was faced unexpectedly with roles of S1P as a first messenger, which subsequently resulted in cloning of its G protein-coupled receptors, S1P1–5. The molecular identification of S1P receptors opened up a new avenue for pathophysiological research on this lipid mediator. Cellular and molecular in vitro studies and in vivo studies on gene deficient mice have elucidated cellular signaling pathways and the pathophysiological meanings of S1P receptors. Another unexpected finding that fingolimod (FTY720) modulates S1P receptors accelerated drug discovery in this field. Fingolimod was approved as a first-in-class, orally active drug for relapsing multiple sclerosis in 2010, and its applications in other disease conditions are currently under clinical trials. In addition, more selective S1P receptor modulators with better pharmacokinetic profiles and fewer side effects are under development. Some of them are being clinically tested in the contexts of multiple sclerosis and other autoimmune and inflammatory disorders, such as, psoriasis, Crohn’s disease, ulcerative colitis, polymyositis, dermatomyositis, liver failure, renal failure, acute stroke, and transplant rejection. In this review, the authors discuss the state of the art regarding the status of drug discovery efforts targeting S1P receptors and place emphasis on potential clinical applications.
Collapse
Affiliation(s)
- Soo-Jin Park
- Molecular Inflammation Research Center for Aging Intervention (MRCA) and College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Dong-Soon Im
- Molecular Inflammation Research Center for Aging Intervention (MRCA) and College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| |
Collapse
|
23
|
Blankenbach KV, Schwalm S, Pfeilschifter J, Meyer Zu Heringdorf D. Sphingosine-1-Phosphate Receptor-2 Antagonists: Therapeutic Potential and Potential Risks. Front Pharmacol 2016; 7:167. [PMID: 27445808 PMCID: PMC4914510 DOI: 10.3389/fphar.2016.00167] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 06/03/2016] [Indexed: 12/26/2022] Open
Abstract
The sphingosine-1-phosphate (S1P) signaling system with its specific G-protein-coupled S1P receptors, the enzymes of S1P metabolism and the S1P transporters, offers a multitude of promising targets for drug development. Until today, drug development in this area has nearly exclusively focused on (functional) antagonists at the S1P1 receptor, which cause a unique phenotype of immunomodulation. Accordingly, the first-in class S1P1 receptor modulator, fingolimod, has been approved for the treatment of relapsing-remitting multiple sclerosis, and novel S1P1 receptor (functional) antagonists are being developed for autoimmune and inflammatory diseases such as psoriasis, inflammatory bowel disease, lupus erythematodes, or polymyositis. Besides the S1P1 receptor, also S1P2 and S1P3 are widely expressed and regulate many diverse functions throughout the body. The S1P2 receptor, in particular, often exerts cellular functions which are opposed to the functions of the S1P1 receptor. As a consequence, antagonists at the S1P2 receptor have the potential to be useful in a contrasting context and different areas of indication compared to S1P1 antagonists. The present review will focus on the therapeutic potential of S1P2 receptor antagonists and discuss their opportunities as well as their potential risks. Open questions and areas which require further investigations will be emphasized in particular.
Collapse
Affiliation(s)
- Kira V Blankenbach
- Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Johann Wolfgang Goethe-Universität Frankfurt am Main, Germany
| | - Stephanie Schwalm
- Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Johann Wolfgang Goethe-Universität Frankfurt am Main, Germany
| | - Josef Pfeilschifter
- Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Johann Wolfgang Goethe-Universität Frankfurt am Main, Germany
| | - Dagmar Meyer Zu Heringdorf
- Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Johann Wolfgang Goethe-Universität Frankfurt am Main, Germany
| |
Collapse
|
24
|
Kitada Y, Kajita K, Taguchi K, Mori I, Yamauchi M, Ikeda T, Kawashima M, Asano M, Kajita T, Ishizuka T, Banno Y, Kojima I, Chun J, Kamata S, Ishii I, Morita H. Blockade of Sphingosine 1-Phosphate Receptor 2 Signaling Attenuates High-Fat Diet-Induced Adipocyte Hypertrophy and Systemic Glucose Intolerance in Mice. Endocrinology 2016; 157:1839-51. [PMID: 26943364 PMCID: PMC4870879 DOI: 10.1210/en.2015-1768] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Sphingosine 1-phosphate (S1P) is known to regulate insulin resistance in hepatocytes, skeletal muscle cells, and pancreatic β-cells. Among its 5 cognate receptors (S1pr1-S1pr5), S1P seems to counteract insulin signaling and confer insulin resistance via S1pr2 in these cells. S1P may also regulate insulin resistance in adipocytes, but the S1pr subtype(s) involved remains unknown. Here, we investigated systemic glucose/insulin tolerance and phenotypes of epididymal adipocytes in high-fat diet (HFD)-fed wild-type and S1pr2-deficient (S1pr2(-/-)) mice. Adult S1pr2(-/-) mice displayed smaller body/epididymal fat tissue weights, but the differences became negligible after 4 weeks with HFD. However, HFD-fed S1pr2(-/-) mice displayed better scores in glucose/insulin tolerance tests and had smaller epididymal adipocytes that expressed higher levels of proliferating cell nuclear antigen than wild-type mice. Next, proliferation/differentiation of 3T3-L1 and 3T3-F442A preadipocytes were examined in the presence of various S1pr antagonists: JTE-013 (S1pr2 antagonist), VPC-23019 (S1pr1/S1pr3 antagonist), and CYM-50358 (S1pr4 antagonist). S1P or JTE-013 treatment of 3T3-L1 preadipocytes potently activated their proliferation and Erk phosphorylation, whereas VPC-23019 inhibited both of these processes, and CYM-50358 had no effects. In contrast, S1P or JTE-013 treatment inhibited adipogenic differentiation of 3T3-F442A preadipocytes, whereas VPC-23019 activated it. The small interfering RNA knockdown of S1pr2 promoted proliferation and inhibited differentiation of 3T3-F442A preadipocytes, whereas that of S1pr1 acted oppositely. Moreover, oral JTE-013 administration improved glucose tolerance/insulin sensitivity in ob/ob mice. Taken together, S1pr2 blockade induced proliferation but suppressed differentiation of (pre)adipocytes both in vivo and in vitro, highlighting a novel therapeutic approach for obesity/type 2 diabetes.
Collapse
Affiliation(s)
- Yoshihiko Kitada
- Department of General Internal Medicine (Y.K., K.K., K.T., I.M., M.Y., T.Ik., M.K., M.A., T.K., H.M.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of General Internal Medicine and Rheumatology (T.Is.), Gifu Municipal Hospital, Gifu 500-8513, Japan; Department of Dermatology (Y.B.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Laboratory of Cell Physiology (I.K.), Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan; Molecular and Cellular Neuroscience Department (J.C.), Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California 92037; and Department of Biochemistry (S.K., I.I.), Keio University Graduate School of Pharmaceutical Sciences, Tokyo 105-8512, Japan
| | - Kazuo Kajita
- Department of General Internal Medicine (Y.K., K.K., K.T., I.M., M.Y., T.Ik., M.K., M.A., T.K., H.M.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of General Internal Medicine and Rheumatology (T.Is.), Gifu Municipal Hospital, Gifu 500-8513, Japan; Department of Dermatology (Y.B.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Laboratory of Cell Physiology (I.K.), Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan; Molecular and Cellular Neuroscience Department (J.C.), Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California 92037; and Department of Biochemistry (S.K., I.I.), Keio University Graduate School of Pharmaceutical Sciences, Tokyo 105-8512, Japan
| | - Koichiro Taguchi
- Department of General Internal Medicine (Y.K., K.K., K.T., I.M., M.Y., T.Ik., M.K., M.A., T.K., H.M.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of General Internal Medicine and Rheumatology (T.Is.), Gifu Municipal Hospital, Gifu 500-8513, Japan; Department of Dermatology (Y.B.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Laboratory of Cell Physiology (I.K.), Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan; Molecular and Cellular Neuroscience Department (J.C.), Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California 92037; and Department of Biochemistry (S.K., I.I.), Keio University Graduate School of Pharmaceutical Sciences, Tokyo 105-8512, Japan
| | - Ichiro Mori
- Department of General Internal Medicine (Y.K., K.K., K.T., I.M., M.Y., T.Ik., M.K., M.A., T.K., H.M.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of General Internal Medicine and Rheumatology (T.Is.), Gifu Municipal Hospital, Gifu 500-8513, Japan; Department of Dermatology (Y.B.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Laboratory of Cell Physiology (I.K.), Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan; Molecular and Cellular Neuroscience Department (J.C.), Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California 92037; and Department of Biochemistry (S.K., I.I.), Keio University Graduate School of Pharmaceutical Sciences, Tokyo 105-8512, Japan
| | - Masahiro Yamauchi
- Department of General Internal Medicine (Y.K., K.K., K.T., I.M., M.Y., T.Ik., M.K., M.A., T.K., H.M.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of General Internal Medicine and Rheumatology (T.Is.), Gifu Municipal Hospital, Gifu 500-8513, Japan; Department of Dermatology (Y.B.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Laboratory of Cell Physiology (I.K.), Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan; Molecular and Cellular Neuroscience Department (J.C.), Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California 92037; and Department of Biochemistry (S.K., I.I.), Keio University Graduate School of Pharmaceutical Sciences, Tokyo 105-8512, Japan
| | - Takahide Ikeda
- Department of General Internal Medicine (Y.K., K.K., K.T., I.M., M.Y., T.Ik., M.K., M.A., T.K., H.M.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of General Internal Medicine and Rheumatology (T.Is.), Gifu Municipal Hospital, Gifu 500-8513, Japan; Department of Dermatology (Y.B.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Laboratory of Cell Physiology (I.K.), Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan; Molecular and Cellular Neuroscience Department (J.C.), Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California 92037; and Department of Biochemistry (S.K., I.I.), Keio University Graduate School of Pharmaceutical Sciences, Tokyo 105-8512, Japan
| | - Mikako Kawashima
- Department of General Internal Medicine (Y.K., K.K., K.T., I.M., M.Y., T.Ik., M.K., M.A., T.K., H.M.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of General Internal Medicine and Rheumatology (T.Is.), Gifu Municipal Hospital, Gifu 500-8513, Japan; Department of Dermatology (Y.B.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Laboratory of Cell Physiology (I.K.), Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan; Molecular and Cellular Neuroscience Department (J.C.), Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California 92037; and Department of Biochemistry (S.K., I.I.), Keio University Graduate School of Pharmaceutical Sciences, Tokyo 105-8512, Japan
| | - Motochika Asano
- Department of General Internal Medicine (Y.K., K.K., K.T., I.M., M.Y., T.Ik., M.K., M.A., T.K., H.M.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of General Internal Medicine and Rheumatology (T.Is.), Gifu Municipal Hospital, Gifu 500-8513, Japan; Department of Dermatology (Y.B.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Laboratory of Cell Physiology (I.K.), Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan; Molecular and Cellular Neuroscience Department (J.C.), Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California 92037; and Department of Biochemistry (S.K., I.I.), Keio University Graduate School of Pharmaceutical Sciences, Tokyo 105-8512, Japan
| | - Toshiko Kajita
- Department of General Internal Medicine (Y.K., K.K., K.T., I.M., M.Y., T.Ik., M.K., M.A., T.K., H.M.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of General Internal Medicine and Rheumatology (T.Is.), Gifu Municipal Hospital, Gifu 500-8513, Japan; Department of Dermatology (Y.B.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Laboratory of Cell Physiology (I.K.), Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan; Molecular and Cellular Neuroscience Department (J.C.), Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California 92037; and Department of Biochemistry (S.K., I.I.), Keio University Graduate School of Pharmaceutical Sciences, Tokyo 105-8512, Japan
| | - Tatsuo Ishizuka
- Department of General Internal Medicine (Y.K., K.K., K.T., I.M., M.Y., T.Ik., M.K., M.A., T.K., H.M.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of General Internal Medicine and Rheumatology (T.Is.), Gifu Municipal Hospital, Gifu 500-8513, Japan; Department of Dermatology (Y.B.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Laboratory of Cell Physiology (I.K.), Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan; Molecular and Cellular Neuroscience Department (J.C.), Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California 92037; and Department of Biochemistry (S.K., I.I.), Keio University Graduate School of Pharmaceutical Sciences, Tokyo 105-8512, Japan
| | - Yoshiko Banno
- Department of General Internal Medicine (Y.K., K.K., K.T., I.M., M.Y., T.Ik., M.K., M.A., T.K., H.M.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of General Internal Medicine and Rheumatology (T.Is.), Gifu Municipal Hospital, Gifu 500-8513, Japan; Department of Dermatology (Y.B.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Laboratory of Cell Physiology (I.K.), Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan; Molecular and Cellular Neuroscience Department (J.C.), Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California 92037; and Department of Biochemistry (S.K., I.I.), Keio University Graduate School of Pharmaceutical Sciences, Tokyo 105-8512, Japan
| | - Itaru Kojima
- Department of General Internal Medicine (Y.K., K.K., K.T., I.M., M.Y., T.Ik., M.K., M.A., T.K., H.M.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of General Internal Medicine and Rheumatology (T.Is.), Gifu Municipal Hospital, Gifu 500-8513, Japan; Department of Dermatology (Y.B.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Laboratory of Cell Physiology (I.K.), Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan; Molecular and Cellular Neuroscience Department (J.C.), Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California 92037; and Department of Biochemistry (S.K., I.I.), Keio University Graduate School of Pharmaceutical Sciences, Tokyo 105-8512, Japan
| | - Jerold Chun
- Department of General Internal Medicine (Y.K., K.K., K.T., I.M., M.Y., T.Ik., M.K., M.A., T.K., H.M.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of General Internal Medicine and Rheumatology (T.Is.), Gifu Municipal Hospital, Gifu 500-8513, Japan; Department of Dermatology (Y.B.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Laboratory of Cell Physiology (I.K.), Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan; Molecular and Cellular Neuroscience Department (J.C.), Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California 92037; and Department of Biochemistry (S.K., I.I.), Keio University Graduate School of Pharmaceutical Sciences, Tokyo 105-8512, Japan
| | - Shotaro Kamata
- Department of General Internal Medicine (Y.K., K.K., K.T., I.M., M.Y., T.Ik., M.K., M.A., T.K., H.M.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of General Internal Medicine and Rheumatology (T.Is.), Gifu Municipal Hospital, Gifu 500-8513, Japan; Department of Dermatology (Y.B.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Laboratory of Cell Physiology (I.K.), Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan; Molecular and Cellular Neuroscience Department (J.C.), Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California 92037; and Department of Biochemistry (S.K., I.I.), Keio University Graduate School of Pharmaceutical Sciences, Tokyo 105-8512, Japan
| | - Isao Ishii
- Department of General Internal Medicine (Y.K., K.K., K.T., I.M., M.Y., T.Ik., M.K., M.A., T.K., H.M.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of General Internal Medicine and Rheumatology (T.Is.), Gifu Municipal Hospital, Gifu 500-8513, Japan; Department of Dermatology (Y.B.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Laboratory of Cell Physiology (I.K.), Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan; Molecular and Cellular Neuroscience Department (J.C.), Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California 92037; and Department of Biochemistry (S.K., I.I.), Keio University Graduate School of Pharmaceutical Sciences, Tokyo 105-8512, Japan
| | - Hiroyuki Morita
- Department of General Internal Medicine (Y.K., K.K., K.T., I.M., M.Y., T.Ik., M.K., M.A., T.K., H.M.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of General Internal Medicine and Rheumatology (T.Is.), Gifu Municipal Hospital, Gifu 500-8513, Japan; Department of Dermatology (Y.B.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Laboratory of Cell Physiology (I.K.), Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan; Molecular and Cellular Neuroscience Department (J.C.), Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California 92037; and Department of Biochemistry (S.K., I.I.), Keio University Graduate School of Pharmaceutical Sciences, Tokyo 105-8512, Japan
| |
Collapse
|
25
|
Zhou F, Zheng T, Abdel-Halim ES, Jiang L, Zhu JJ. A multifunctional core–shell nanoplatform for enhanced cancer cell apoptosis and targeted chemotherapy. J Mater Chem B 2016; 4:2887-2894. [DOI: 10.1039/c6tb00438e] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
A novel multifunctional nanoplatform was designed based on the combination of silver nanoparticles (AgNPs) with nucleolin-targeted and doxorubicin (Dox)-loaded manganese dioxide (MnO2) nanosheets to induce enhanced cancer cell apoptosis and targeted chemotherapy.
Collapse
Affiliation(s)
- Fei Zhou
- State Key Laboratory of Analytical Chemistry for Life Science
- School of Chemistry and Chemical Engineering
- Nanjing University
- Nanjing 210093
- P. R. China
| | - Tingting Zheng
- State Key Laboratory of Analytical Chemistry for Life Science
- School of Chemistry and Chemical Engineering
- Nanjing University
- Nanjing 210093
- P. R. China
| | - E. S. Abdel-Halim
- Chemistry Department
- College of Science
- King Saud University
- Riyadh 11451
- Kingdom of Saudi Arabia
| | - Liping Jiang
- State Key Laboratory of Analytical Chemistry for Life Science
- School of Chemistry and Chemical Engineering
- Nanjing University
- Nanjing 210093
- P. R. China
| | - Jun-Jie Zhu
- State Key Laboratory of Analytical Chemistry for Life Science
- School of Chemistry and Chemical Engineering
- Nanjing University
- Nanjing 210093
- P. R. China
| |
Collapse
|
26
|
Japtok L, Schmitz EI, Fayyaz S, Krämer S, Hsu LJ, Kleuser B. Sphingosine 1-phosphate counteracts insulin signaling in pancreatic β-cells via the sphingosine 1-phosphate receptor subtype 2. FASEB J 2015; 29:3357-69. [PMID: 25911610 DOI: 10.1096/fj.14-263194] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 04/16/2015] [Indexed: 01/04/2023]
Abstract
Glucolipotoxic stress has been identified as a key player in the progression of pancreatic β-cell dysfunction contributing to insulin resistance and the development of type 2 diabetes mellitus (T2D). It has been suggested that bioactive lipid intermediates, formed under lipotoxic conditions, are involved in these processes. Here, we show that sphingosine 1-phosphate (S1P) levels are not only increased in palmitate-stimulated pancreatic β-cells but also regulate β-cell homeostasis in a divergent manner. Although S1P possesses a prosurvival effect in β-cells, an enhanced level of the sphingolipid antagonizes insulin-mediated cell growth and survival via the sphingosine 1-phosphate receptor subtype 2 (S1P2) followed by an inhibition of Akt-signaling. In an attempt to investigate the role of the S1P/S1P2 axis in vivo, the New Zealand obese (NZO) diabetic mouse model, characterized by β-cell loss under high-fat diet (HFD) conditions, was used. The occurrence of T2D was accompanied by an increase of plasma S1P levels. To examine whether S1P contributes to the morphologic changes of islets via S1P2, the receptor antagonist JTE-013 was administered. Most interestingly, JTE-013 rescued β-cell damage clearly indicating an important role of the S1P2 in β-cell homeostasis. Therefore, the present study provides a new therapeutic strategy to diminish β-cell dysfunction and the development of T2D.
Collapse
Affiliation(s)
- Lukasz Japtok
- *Faculty of Mathematics and Natural Science, Institute of Nutritional Science, Department of Toxicology, University of Potsdam, Potsdam, Germany; German Institute of Human Nutrition, Max Rubner Laboratory, Nuthetal, Germany; and Lpath Incorporated, San Diego, California, USA
| | - Elisabeth I Schmitz
- *Faculty of Mathematics and Natural Science, Institute of Nutritional Science, Department of Toxicology, University of Potsdam, Potsdam, Germany; German Institute of Human Nutrition, Max Rubner Laboratory, Nuthetal, Germany; and Lpath Incorporated, San Diego, California, USA
| | - Susann Fayyaz
- *Faculty of Mathematics and Natural Science, Institute of Nutritional Science, Department of Toxicology, University of Potsdam, Potsdam, Germany; German Institute of Human Nutrition, Max Rubner Laboratory, Nuthetal, Germany; and Lpath Incorporated, San Diego, California, USA
| | - Stephanie Krämer
- *Faculty of Mathematics and Natural Science, Institute of Nutritional Science, Department of Toxicology, University of Potsdam, Potsdam, Germany; German Institute of Human Nutrition, Max Rubner Laboratory, Nuthetal, Germany; and Lpath Incorporated, San Diego, California, USA
| | - Leigh J Hsu
- *Faculty of Mathematics and Natural Science, Institute of Nutritional Science, Department of Toxicology, University of Potsdam, Potsdam, Germany; German Institute of Human Nutrition, Max Rubner Laboratory, Nuthetal, Germany; and Lpath Incorporated, San Diego, California, USA
| | - Burkhard Kleuser
- *Faculty of Mathematics and Natural Science, Institute of Nutritional Science, Department of Toxicology, University of Potsdam, Potsdam, Germany; German Institute of Human Nutrition, Max Rubner Laboratory, Nuthetal, Germany; and Lpath Incorporated, San Diego, California, USA
| |
Collapse
|
27
|
Bellini L, Campana M, Mahfouz R, Carlier A, Véret J, Magnan C, Hajduch E, Le Stunff H. Targeting sphingolipid metabolism in the treatment of obesity/type 2 diabetes. Expert Opin Ther Targets 2015; 19:1037-50. [PMID: 25814122 DOI: 10.1517/14728222.2015.1028359] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
INTRODUCTION Obesity is a major factor that is linked to the development of type 2 diabetes (T2D). Excess circulating fatty acids (FAs), which characterize obesity, induce insulin resistance, steatosis, β cells dysfunction and apoptosis. These deleterious effects have been defined as lipotoxicity. AREAS COVERED FAs are metabolized to different lipid species, including ceramides which play a crucial role in lipotoxicity. The action of ceramides on tissues, such as muscle, liver, adipose tissue and pancreatic β cells, during the development of T2D will also be reviewed. In addition, the potential antagonist action of other sphingolipids, namely sphingoid base phosphates, on lipotoxicity in skeletal muscle and β cells will be addressed. EXPERT OPINION Ceramide is a critical mediator to the development of T2D linked to obesity. Targeting proteins involved in ceramide's deleterious action has not been possible due to their involvement in many other intracellular signaling pathways. A possible means of counteracting ceramide action would be to prevent the accumulation of the specific ceramide species involved in both insulin resistance and β-cell apoptosis/dysfunction. Another possibility would be to adjust the dynamic balance between ceramide and sphingoid base phosphate, both known to display opposing properties on the development of T2D-linked obesity.
Collapse
Affiliation(s)
- Lara Bellini
- Université PARIS-DIDEROT (7), Unité Biologie Fonctionnelle et Adaptative - UMR CNRS 8251, Équipe Régulation de la glycémie par le système nerveux central (REGLYS) , 4, rue Marie-Andrée Lagroua Weill-Halle, 75205 PARIS Cedex 13 , France +01 57 27 77 97 ; +01 57 27 77 96 ;
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Switching the sphingolipid rheostat in the treatment of diabetes and cancer comorbidity from a problem to an advantage. BIOMED RESEARCH INTERNATIONAL 2015; 2015:165105. [PMID: 25866760 PMCID: PMC4383402 DOI: 10.1155/2015/165105] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 10/16/2014] [Indexed: 12/11/2022]
Abstract
Cancer and diabetes are among the most common diseases in western societies. Epidemiological studies have shown that diabetic patients have a significantly higher risk of developing a number of different types of cancers and that individuals with comorbidity (cancer and diabetes/prediabetes) have a poorer prognosis relative to nondiabetic cancer patients. The increasing frequency of comorbidity of cancer and diabetes mellitus, mainly type 2 diabetes, has driven the development of therapeutic interventions that target both disease states. There is strong evidence to suggest that balancing the sphingolipid rheostat, ceramide--sphingosine--sphingosine-1-phosphate (S1P) is crucial in the prevention of diabetes and cancer and sphingosine kinase/S1P modulators are currently under development for the treatment of cancer and diabetes. This paper will highlight some of the complexities inherent in the use of the emerging sphingosine kinase/S1P modulators in the treatment of comorbidity of diabetes and cancer.
Collapse
|
29
|
Fernández-Velasco M, Ruiz-Hurtado G, Gómez AM, Rueda A. Ca(2+) handling alterations and vascular dysfunction in diabetes. Cell Calcium 2014; 56:397-407. [PMID: 25218935 DOI: 10.1016/j.ceca.2014.08.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 07/30/2014] [Accepted: 08/07/2014] [Indexed: 12/12/2022]
Abstract
More than 65% of patients with diabetes mellitus die from cardiovascular disease or stroke. Hyperglycemia, due to either reduced insulin secretion or reduced insulin sensitivity, is the hallmark feature of diabetes mellitus. Vascular dysfunction is a distinctive phenotype found in both types of diabetes and could be responsible for the high incidence of stroke, heart attack, and organ damage in diabetic patients. In addition to well-documented endothelial dysfunction, Ca(2+) handling alterations in vascular smooth muscle cells (VSMCs) play a key role in the development and progression of vascular complications in diabetes. VSMCs provide not only structural integrity to the vessels but also control myogenic arterial tone and systemic blood pressure through global and local Ca(2+) signaling. The Ca(2+) signalosome of VSMCs is integrated by an extensive number of Ca(2+) handling proteins (i.e. channels, pumps, exchangers) and related signal transduction components, whose function is modulated by endothelial effectors. This review summarizes recent findings concerning alterations in endothelium and VSMC Ca(2+) signaling proteins that may contribute to the vascular dysfunction found in the diabetic condition.
Collapse
Affiliation(s)
| | - Gema Ruiz-Hurtado
- Unidad de Hipertensión, Instituto de Investigación imas12, Hospital 12 de Octubre, Madrid, Spain; Instituto Pluridisciplinar, Facultad de Farmacia, Universidad Complutense de Madrid, Spain
| | - Ana M Gómez
- Inserm, UMR S769, Faculté de Pharmacie, Université Paris Sud, Labex LERMIT, DHU TORINO, Châtenay-Malabry, France
| | - Angélica Rueda
- Departamento de Bioquímica, Centro de Investigación y de Estudios Avanzados del IPN, México City, Mexico.
| |
Collapse
|
30
|
Roles of Sphingolipid Metabolism in Pancreatic β Cell Dysfunction Induced by Lipotoxicity. J Clin Med 2014; 3:646-62. [PMID: 26237395 PMCID: PMC4449690 DOI: 10.3390/jcm3020646] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 04/10/2014] [Accepted: 04/11/2014] [Indexed: 12/31/2022] Open
Abstract
Pancreatic β cells secrete insulin in order to maintain glucose homeostasis. However, various environmental stresses such as obesity have been shown to induce loss of secretory responsiveness in pancreatic β cells and pancreatic β cell apoptosis which can favor the development of type 2 diabetes (T2D). Indeed, elevated levels of free fatty acids (FFAs) have been shown to induce β cell apoptosis. Importantly, the chronic adverse effects of FFAs on β cell function and viability are potentiated in the presence of hyperglycaemia, a phenomenon that has been termed gluco-lipotoxicity. The molecular mechanisms underlying the pathogenesis of gluco-lipotoxicity in pancreatic β cells are not completely understood. Recent studies have shown that sphingolipid metabolism plays a key role in gluco-lipotoxicity induced apoptosis and loss of function of pancreatic β cells. The present review focuses on how the two main sphingolipid mediators, ceramides and sphingoid base-1-phosphates, regulate the deleterious effects of gluco-lipotoxicity on pancreatic β cells. The review highlights the role of a sphingolipid biostat on the dysregulation of β cell fate and function induced by gluco-lipotoxicity, offering the possibility of new therapeutic targets to prevent the onset of T2D.
Collapse
|
31
|
Fayyaz S, Henkel J, Japtok L, Krämer S, Damm G, Seehofer D, Püschel GP, Kleuser B. Involvement of sphingosine 1-phosphate in palmitate-induced insulin resistance of hepatocytes via the S1P2 receptor subtype. Diabetologia 2014; 57:373-82. [PMID: 24292566 DOI: 10.1007/s00125-013-3123-6] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 11/06/2013] [Indexed: 02/06/2023]
Abstract
AIMS/HYPOTHESIS Enhanced plasma levels of NEFA have been shown to induce hepatic insulin resistance, which contributes to the development of type 2 diabetes. Indeed, sphingolipids can be formed via a de novo pathway from the saturated fatty acid palmitate and the amino acid serine. Besides ceramides, sphingosine 1-phosphate (S1P) has been identified as a major bioactive lipid mediator. Therefore, our aim was to investigate the generation and function of S1P in hepatic insulin resistance. METHODS The incorporation of palmitate into sphingolipids was performed by rapid-resolution liquid chromatography-MS/MS in primary human and rat hepatocytes. The influence of S1P and the involvement of S1P receptors in hepatic insulin resistance was examined in human and rat hepatocytes, as well as in New Zealand obese (NZO) mice. RESULTS Palmitate induced an impressive formation of extra- and intracellular S1P in rat and human hepatocytes. An elevation of hepatic S1P levels was observed in NZO mice fed a high-fat diet. Once generated, S1P was able, similarly to palmitate, to counteract insulin signalling. The inhibitory effect of S1P was abolished in the presence of the S1P2 receptor antagonist JTE-013 both in vitro and in vivo. In agreement with this, the immunomodulator FTY720-phosphate, which binds to all S1P receptors except S1P2, was not able to inhibit insulin signalling. CONCLUSIONS/INTERPRETATION These data indicate that palmitate is metabolised by hepatocytes to S1P, which acts via stimulation of the S1P2 receptor to impair insulin signalling. In particular, S1P2 inhibition could be considered as a novel therapeutic target for the treatment of insulin resistance.
Collapse
Affiliation(s)
- Susann Fayyaz
- Faculty of Mathematics and Natural Science, Institute of Nutritional Science, Department of Toxicology, University of Potsdam, Arthur-Scheunert Allee 114-116, 14558, Nuthetal, Potsdam, Germany
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Adada M, Canals D, Hannun YA, Obeid LM. Sphingosine-1-phosphate receptor 2. FEBS J 2013; 280:6354-66. [PMID: 23879641 DOI: 10.1111/febs.12446] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Revised: 07/15/2013] [Accepted: 07/22/2013] [Indexed: 12/15/2022]
Abstract
Sphingosine-1-phosphate (S1P) is a potent bioactive sphingolipid involved in cell proliferation, angiogenesis, inflammation and malignant transformation among other functions. S1P acts either directly on intracellular targets or activates G protein-coupled receptors, specifically five S1P receptors (S1PRs). The identified S1PRs differ in cellular and tissue distribution, and each is coupled to specific G proteins, which mediate unique functions. Here, we describe functional characteristics of all five receptors, emphasizing S1PR2, which is critical in the immune, nervous, metabolic, cardiovascular, musculoskeletal, and renal systems. This review also describes the role of this receptor in tumor growth and metastasis and suggests potential therapeutic avenues that exploit S1PR2.
Collapse
Affiliation(s)
- Mohamad Adada
- Department of Medicine, Stony Brook University, NY, USA
| | | | | | | |
Collapse
|
33
|
Choi JW, Chun J. Lysophospholipids and their receptors in the central nervous system. Biochim Biophys Acta Mol Cell Biol Lipids 2012; 1831:20-32. [PMID: 22884303 DOI: 10.1016/j.bbalip.2012.07.015] [Citation(s) in RCA: 194] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Revised: 07/17/2012] [Accepted: 07/18/2012] [Indexed: 02/05/2023]
Abstract
Lysophosphatidic acid (LPA) and sphingosine 1-phosphate (S1P), two of the best-studied lysophospholipids, are known to influence diverse biological events, including organismal development as well as function and pathogenesis within multiple organ systems. These functional roles are due to a family of at least 11 G protein-coupled receptors (GPCRs), named LPA(1-6) and S1P(1-5), which are widely distributed throughout the body and that activate multiple effector pathways initiated by a range of heterotrimeric G proteins including G(i/o), G(12/13), G(q) and G(s), with actual activation dependent on receptor subtypes. In the central nervous system (CNS), a major locus for these signaling pathways, LPA and S1P have been shown to influence myriad responses in neurons and glial cell types through their cognate receptors. These receptor-mediated activities can contribute to disease pathogenesis and have therapeutic relevance to human CNS disorders as demonstrated for multiple sclerosis (MS) and possibly others that include congenital hydrocephalus, ischemic stroke, neurotrauma, neuropsychiatric disorders, developmental disorders, seizures, hearing loss, and Sandhoff disease, based upon the experimental literature. In particular, FTY720 (fingolimod, Gilenya, Novartis Pharma, AG) that becomes an analog of S1P upon phosphorylation, was approved by the FDA in 2010 as a first oral treatment for MS, validating this class of receptors as medicinal targets. This review will provide an overview and update on the biological functions of LPA and S1P signaling in the CNS, with a focus on results from studies using genetic null mutants for LPA and S1P receptors. This article is part of a Special Issue entitled Advances in Lysophospholipid Research.
Collapse
Affiliation(s)
- Ji Woong Choi
- Department of Molecular Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | |
Collapse
|
34
|
Shea BS, Tager AM. Sphingolipid regulation of tissue fibrosis. Open Rheumatol J 2012; 6:123-9. [PMID: 22802910 PMCID: PMC3395890 DOI: 10.2174/1874312901206010123] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2012] [Revised: 03/27/2012] [Accepted: 04/04/2012] [Indexed: 01/08/2023] Open
Abstract
Bioactive sphingolipids, such as sphingosine 1-phosphate (S1P), dihydrosphingosine 1-phosphate (dhS1P) and ceramide, regulate a diverse array of cellular processes. Many of these processes are important components of wound-healing responses to tissue injury, including cellular apoptosis, vascular leak, fibroblast migration, and TGF-β signaling. Since over-exuberant or aberrant wound-healing responses to repetitive injury have been implicated in the pathogenesis of tissue fibrosis, these signaling sphingolipids have the potential to influence the development and progression of fibrotic diseases. Here we review accumulating in vitro and in vivo data indicating that these lipid mediators can in fact influence fibrogenesis in numerous organ systems, including the lungs, skin, liver, heart, and eye. Targeting these lipids, their receptors, or the enzymes involved in their metabolism consequently now appears to hold great promise for the development of novel therapies for fibrotic diseases.
Collapse
Affiliation(s)
- Barry S Shea
- Pulmonary and Critical Care Unit, and Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA 02114, USA
| | | |
Collapse
|
35
|
Li C, Chi XX, Xie W, Strong JA, Zhang JM, Nicol GD. Sphingosine 1-phosphate receptor 2 antagonist JTE-013 increases the excitability of sensory neurons independently of the receptor. J Neurophysiol 2012; 108:1473-83. [PMID: 22673325 DOI: 10.1152/jn.00825.2011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Previously we demonstrated that sphingosine 1-phosphate receptor 1 (S1PR(1)) played a prominent, but not exclusive, role in enhancing the excitability of small-diameter sensory neurons, suggesting that other S1PRs can modulate neuronal excitability. To examine the potential role of S1PR(2) in regulating neuronal excitability we used the established selective antagonist of S1PR(2), JTE-013. Here we report that exposure to JTE-013 alone produced a significant increase in excitability in a time- and concentration-dependent manner in 70-80% of recorded neurons. Internal perfusion of sensory neurons with guanosine 5'-O-(2-thiodiphosphate) (GDP-β-S) via the recording pipette inhibited the sensitization produced by JTE-013 as well as prostaglandin E(2). Pretreatment with pertussis toxin or the selective S1PR(1) antagonist W146 blocked the sensitization produced by JTE-013. These results indicate that JTE-013 might act as an agonist at other G protein-coupled receptors. In neurons that were sensitized by JTE-013, single-cell RT-PCR studies demonstrated that these neurons did not express the mRNA for S1PR(2). In behavioral studies, injection of JTE-013 into the rat's hindpaw produced a significant increase in the mechanical sensitivity in the ipsilateral, but not contralateral, paw. Injection of JTE-013 did not affect the withdrawal latency to thermal stimulation. Thus JTE-013 augments neuronal excitability independently of S1PR(2) by unknown mechanisms that may involve activation of other G protein-coupled receptors such as S1PR(1). Clearly, further studies are warranted to establish the causal nature of this increased sensitivity, and future studies of neuronal function using JTE-013 should be interpreted with caution.
Collapse
Affiliation(s)
- Chao Li
- Medical Neuroscience Program, School of Medicine, Indiana University, Indianapolis, IN, USA
| | | | | | | | | | | |
Collapse
|
36
|
Mutoh T, Rivera R, Chun J. Insights into the pharmacological relevance of lysophospholipid receptors. Br J Pharmacol 2012; 165:829-44. [PMID: 21838759 PMCID: PMC3312481 DOI: 10.1111/j.1476-5381.2011.01622.x] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2011] [Revised: 07/22/2011] [Accepted: 07/23/2011] [Indexed: 12/22/2022] Open
Abstract
The discovery of lysophospholipid (LP) 7-transmembrane, G protein-coupled receptors (GPCRs) that began in the 1990s, together with research into the functional roles of the major LPs known as lysophosphatidic acid (LPA) and sphingosine 1-phosphate (S1P), have opened new research avenues into their biological processes and mechanisms. Major examples of LP signalling effects include embryogenesis, nervous system development, vascular development, uterine implantation, immune cell trafficking, and inflammatory reactions. LP signalling also influences the pathophysiology of many diseases including cancer, autoimmune and inflammatory diseases, which indicate that LP receptors may be attractive targets for pharmacological therapies. A key example of such a therapeutic agent is the S1P receptor modulator FTY720, which upon phosphorylation and continued drug exposure, acts as an S1P receptor functional antagonist. This compound (also known as fingolimod or Gilenya) has recently been approved by the FDA for the treatment of relapsing forms of multiple sclerosis. Continued basic and translational research on LP signalling should provide novel insights into both basic biological mechanisms, as well as novel therapeutic approaches to combat a range of human diseases.
Collapse
Affiliation(s)
- Tetsuji Mutoh
- Department of Molecular Biology, Dorris Neuroscience Center, The Scripps Research InstituteLa Jolla, CA, USA
- Gunma Kokusai AcademyGunma, Japan
| | - Richard Rivera
- Department of Molecular Biology, Dorris Neuroscience Center, The Scripps Research InstituteLa Jolla, CA, USA
| | - Jerold Chun
- Department of Molecular Biology, Dorris Neuroscience Center, The Scripps Research InstituteLa Jolla, CA, USA
| |
Collapse
|
37
|
Studer E, Zhou X, Zhao R, Wang Y, Takabe K, Nagahashi M, Pandak WM, Dent P, Spiegel S, Shi R, Xu W, Liu X, Bohdan P, Zhang L, Zhou H, Hylemon PB. Conjugated bile acids activate the sphingosine-1-phosphate receptor 2 in primary rodent hepatocytes. Hepatology 2012; 55:267-76. [PMID: 21932398 PMCID: PMC3245352 DOI: 10.1002/hep.24681] [Citation(s) in RCA: 233] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Accepted: 08/29/2011] [Indexed: 12/15/2022]
Abstract
Bile acids have been shown to be important regulatory molecules for cells in the liver and gastrointestinal tract. They can activate various cell signaling pathways including extracellular regulated kinase (ERK)1/2 and protein kinase B (AKT) as well as the G-protein-coupled receptor (GPCR) membrane-type bile acid receptor (TGR5/M-BAR). Activation of the ERK1/2 and AKT signaling pathways by conjugated bile acids has been reported to be sensitive to pertussis toxin (PTX) and dominant-negative Gα(i) in primary rodent hepatocytes. However, the GPCRs responsible for activation of these pathways have not been identified. Screening GPCRs in the lipid-activated phylogenetic family (expressed in HEK293 cells) identified sphingosine-1-phosphate receptor 2 (S1P(2) ) as being activated by taurocholate (TCA). TCA, taurodeoxycholic acid (TDCA), tauroursodeoxycholic acid (TUDCA), glycocholic acid (GCA), glycodeoxycholic acid (GDCA), and S1P-induced activation of ERK1/2 and AKT were significantly inhibited by JTE-013, a S1P(2) antagonist, in primary rat hepatocytes. JTE-013 significantly inhibited hepatic ERK1/2 and AKT activation as well as short heterodimeric partner (SHP) mRNA induction by TCA in the chronic bile fistula rat. Knockdown of the expression of S1P(2) by a recombinant lentivirus encoding S1P(2) shRNA markedly inhibited the activation of ERK1/2 and AKT by TCA and S1P in rat primary hepatocytes. Primary hepatocytes prepared from S1P(2) knock out (S1P(2) (-/-) ) mice were significantly blunted in the activation of the ERK1/2 and AKT pathways by TCA. Structural modeling of the S1P receptors indicated that only S1P(2) can accommodate TCA binding. In summary, all these data support the hypothesis that conjugated bile acids activate the ERK1/2 and AKT signaling pathways primarily through S1P(2) in primary rodent hepatocytes.
Collapse
Affiliation(s)
- Elaine Studer
- Department of Microbiology and Immunology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298
| | - Xiqiao Zhou
- Department of Microbiology and Immunology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298,Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu, China
| | - Renping Zhao
- Department of Microbiology and Immunology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298,China Pharmaceutical University, Nanjing, China
| | - Yun Wang
- Department of Microbiology and Immunology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298,China Pharmaceutical University, Nanjing, China
| | - Kazuaki Takabe
- Department of Surgery, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298
| | - Masayuki Nagahashi
- Department of Surgery, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298
| | - William M. Pandak
- McGuire Veterans Affairs Medical Center, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298
| | - Paul Dent
- Department of Neurosurgery, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298
| | - Ruihua Shi
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu, China
| | - Weiren Xu
- Tianjin Key Laboratory of Molecular Design and Drug Discovery, Tianjin Institute of Pharmaceutical Research, Tianjin China
| | - Xuyuan Liu
- Tianjin Key Laboratory of Molecular Design and Drug Discovery, Tianjin Institute of Pharmaceutical Research, Tianjin China
| | - Pat Bohdan
- Department of Microbiology and Immunology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298
| | | | - Huiping Zhou
- Department of Microbiology and Immunology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298,McGuire Veterans Affairs Medical Center, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298,Address: To whom correspondence should be addressed: Phillip B. Hylemon, Ph.D., Department of Microbiology and Immunology, Medical College of Virginia Campus-VCU, PO Box 908678, Richmond, VA 23298-0678, Tel. (804) 347-1752; Fax. (804) 828-0676, Or Huiping Zhou, Ph.D, Department of Microbiology and Immunology, Medical College of Virginia Campus-VCU, PO Box 908678, Richmond, VA 23298-0678, Tel. (804)828-6817; Fax. (804) 828-0676,
| | - Phillip B. Hylemon
- Department of Microbiology and Immunology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298,McGuire Veterans Affairs Medical Center, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298,Address: To whom correspondence should be addressed: Phillip B. Hylemon, Ph.D., Department of Microbiology and Immunology, Medical College of Virginia Campus-VCU, PO Box 908678, Richmond, VA 23298-0678, Tel. (804) 347-1752; Fax. (804) 828-0676, Or Huiping Zhou, Ph.D, Department of Microbiology and Immunology, Medical College of Virginia Campus-VCU, PO Box 908678, Richmond, VA 23298-0678, Tel. (804)828-6817; Fax. (804) 828-0676,
| |
Collapse
|
38
|
Akahoshi N, Ishizaki Y, Yasuda H, Murashima YL, Shinba T, Goto K, Himi T, Chun J, Ishii I. Frequent spontaneous seizures followed by spatial working memory/anxiety deficits in mice lacking sphingosine 1-phosphate receptor 2. Epilepsy Behav 2011; 22:659-65. [PMID: 22019019 DOI: 10.1016/j.yebeh.2011.09.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Revised: 08/31/2011] [Accepted: 09/03/2011] [Indexed: 11/16/2022]
Abstract
The diverse physiological effects of sphingosine 1-phosphate (S1P) are mostly mediated by its five cognate G protein-coupled receptors, S1P(1)-S1P(5), which have attracted much attention as future drug targets. To gain insight into S1P(2)-mediated signaling, we analyzed frequent spontaneous seizures in S1P(2)-deficient (S1P(2)(-/-)) mice obtained after several backcrosses onto a C57BL/6N background. Full-time video recording of 120 S1P(2)(-/-) mice identified 420 seizures both day and night between postnatal days 25 and 45, which were accompanied by high-voltage synchronized cortical discharges and a series of typical episodes: wild run, tonic-clonic convulsion, freezing, and, occasionally, death. Nearly 40% of 224 S1P(2)(-/-) mice died after such seizures, while the remaining 60% of the mice survived to adulthood; however, approximately half of the deliveries from S1P(2)(-/-) pregnant mice resulted in neonatal death. In situ hybridization revealed exclusive s1p(2) expression in the hippocampal pyramidal/granular neurons of wild-type mice, and immunohistochemistry/microarray analyses identified enhanced gliosis in the whole hippocampus and its neighboring neocortex in seizure-prone adult S1P(2)(-/-) mice. Seizure-prone adult S1P(2)(-/-) mice displayed impaired spatial working memory in the eight-arm radial maze test and increased anxiety in the elevated plus maze test, whereas their passive avoidance learning memory performance in the step-through test and hippocampal long-term potentiation was indistinguishable from that of wild-type mice. Our findings suggest that blockade of S1P(2) signaling may cause seizures/hippocampal insults and impair some specific central nervous system functions.
Collapse
Affiliation(s)
- Noriyuki Akahoshi
- Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, Gunma, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Sphingosine-1-phosphate signaling and its role in disease. Trends Cell Biol 2011; 22:50-60. [PMID: 22001186 DOI: 10.1016/j.tcb.2011.09.003] [Citation(s) in RCA: 784] [Impact Index Per Article: 60.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Revised: 09/08/2011] [Accepted: 09/09/2011] [Indexed: 02/07/2023]
Abstract
The bioactive sphingolipid metabolite sphingosine-1-phosphate (S1P) is now recognized as a critical regulator of many physiological and pathophysiological processes, including cancer, atherosclerosis, diabetes and osteoporosis. S1P is produced in cells by two sphingosine kinase isoenzymes, SphK1 and SphK2. Many cells secrete S1P, which can then act in an autocrine or paracrine manner. Most of the known actions of S1P are mediated by a family of five specific G protein-coupled receptors. More recently, it was shown that S1P also has important intracellular targets involved in inflammation, cancer and Alzheimer's disease. This suggests that S1P actions are much more complex than previously thought, with important ramifications for development of therapeutics. This review highlights recent advances in our understanding of the mechanisms of action of S1P and its roles in disease.
Collapse
|
40
|
Bautista-Pérez R, Arellano A, Franco M, Osorio H, Coronel I. Sphingosine-1-phosphate induced vasoconstriction is increased in the isolated perfused kidneys of diabetic rats. Diabetes Res Clin Pract 2011; 94:e8-11. [PMID: 21775010 DOI: 10.1016/j.diabres.2011.06.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2011] [Revised: 06/09/2011] [Accepted: 06/26/2011] [Indexed: 11/18/2022]
Abstract
We observed that in isolated perfused rat kidneys, sphingosine-1-phosphate produces S1P(2) receptor-mediated vasoconstriction, and this response increased in kidneys of diabetic rats. These results suggest that the antagonists of S1P(2) receptor may have potential as drugs to control diabetes-induced vascular complications.
Collapse
Affiliation(s)
- Rocio Bautista-Pérez
- Department of Nephrology, Instituto Nacional de Cardiologia Ignacio Chavez, México City, Mexico.
| | | | | | | | | |
Collapse
|
41
|
Pyne NJ, Pyne S. Selectivity and specificity of sphingosine 1-phosphate receptor ligands: "off-targets" or complex pharmacology? Front Pharmacol 2011; 2:26. [PMID: 21687518 PMCID: PMC3108476 DOI: 10.3389/fphar.2011.00026] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2011] [Accepted: 05/20/2011] [Indexed: 01/19/2023] Open
Abstract
A recent perspective published in Frontiers of Pharmacology by Salomone and Waeber (2011) discussed the selectivity and specificity of sphingosine 1-phosphate (S1P) receptor ligands. This perspective surveyed the use of various S1P receptor ligands and attempted to reconcile a number of inconsistencies in the predicted biological outcomes: these were interpreted as “off-target” effects. Therefore the perspective cautioned against the use of these S1P receptor ligands. Here we highlight the complex pharmacology of S1P receptors, which along with “inside-out” signaling might provide an alternative explanation for “off-target” effects.
Collapse
Affiliation(s)
- Nigel J Pyne
- Cell Biology Group, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde Glasgow, UK
| | | |
Collapse
|
42
|
Pyne S, Pyne NJ. Translational aspects of sphingosine 1-phosphate biology. Trends Mol Med 2011; 17:463-72. [PMID: 21514226 DOI: 10.1016/j.molmed.2011.03.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Revised: 02/28/2011] [Accepted: 03/04/2011] [Indexed: 12/16/2022]
Abstract
Sphingosine 1-phosphate (S1P) is a bioactive lipid that has both physiological and pathophysiological roles. It regulates cellular processes such as proliferation, migration, survival and differentiation and affects all organ systems. S1P not only activates S1P-specific receptors to initiate cellular signalling pathways but also directly regulates specific intracellular target proteins. The therapeutic opportunities surrounding S1P signalling are numerous and exemplified by the recent approval of FTY720 (a sphingosine analogue, Gilenya™) for the treatment of relapsing multiple sclerosis. A major focus of research is to develop small-molecule antagonists/agonists/inhibitors that are specific to the different S1P receptor subtypes and the enzymes that regulate S1P levels. This review describes fundamental aspects of S1P biology with an emphasis on the translational potential of intervention therapeutics.
Collapse
Affiliation(s)
- Susan Pyne
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK.
| | | |
Collapse
|
43
|
Roviezzo F, Brancaleone V, De Gruttola L, Vellecco V, Bucci M, D'Agostino B, Cooper D, Sorrentino R, Perretti M, Cirino G. Sphingosine-1-phosphate modulates vascular permeability and cell recruitment in acute inflammation in vivo. J Pharmacol Exp Ther 2011; 337:830-7. [PMID: 21421740 DOI: 10.1124/jpet.111.179168] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The sphingosine kinase (SPK)/sphingosine-1-phosphate (S1P) pathway recently has been associated with a variety of inflammatory-based diseases. The majority of these studies have been performed in vitro. Here, we have addressed the relevance of the SPK/S1P pathway in the acute inflammatory response in vivo by using different well known preclinical animal models. The study has been performed by operating a pharmacological modulation using 1) L-cycloserine and DL-threo-dihydrosphingosine (DTD), S1P synthesis inhibitors or 2) 2-undecyl-thiazolidine-4-carboxylic acid (BML-241) and N-(2,6-dichloro-4-pyridinyl)-2-[1,3-dimethyl-4-(1-methylethyl)-1H-pyrazolo[3,4-b]pyridin-6-yl]-hydrazinecarboxamide (JTE-013), specific S1P(2) and S1P(3) receptor antagonists. After local injection of carrageenan in mouse paw S1P release significantly increases locally and decreases during the resolution phase. Expression of SPKs and S1P(2) and S1P(3) receptors is increased in inflamed tissues. Administration of L-cycloserine or DTD caused a significant anti-inflammatory effect. By using different animal models we have also demonstrated that the SPK/S1P pathway contributes to changes in vascular permeability and promotes cell recruitment. The S1P effect on cell recruitment results is receptor-mediated because both JTE-013 and BML-241 inhibited zymosan-induced cell chemotaxis without effect on vascular leakage. Conversely, changes in vascular permeability involve mainly SPK activity, because compound 48/80-induced vascular leakage was significantly inhibited by DTD. In conclusion, the SPK/S1P pathway is involved in acute inflammation and could represent a valuable therapeutic target for developing a new class of anti-inflammatory drugs.
Collapse
Affiliation(s)
- Fiorentina Roviezzo
- Dipartimento di Farmacologia Sperimentale, Università di Napoli Federico II, Domenico Montesano 49, 80131 Naples, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Kobayashi NR, Hawes SM, Crook JM, Pébay A. G-protein coupled receptors in stem cell self-renewal and differentiation. Stem Cell Rev Rep 2010; 6:351-66. [PMID: 20625855 DOI: 10.1007/s12015-010-9167-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Stem cells have great potential for understanding early development, treating human disease, tissue trauma and early phase drug discovery. The factors that control the regulation of stem cell survival, proliferation, migration and differentiation are still emerging. Some evidence now exists demonstrating the potent effects of various G-protein coupled receptor (GPCR) ligands on the biology of stem cells. This review aims to give an overview of the current knowledge of the regulation of embryonic and somatic stem cell maintenance and differentiation by GPCR ligands.
Collapse
|