1
|
Wang J, Zheng G, Wang L, Meng L, Ren J, Shang L, Li D, Bao Y. Dysregulation of sphingolipid metabolism in pain. Front Pharmacol 2024; 15:1337150. [PMID: 38523645 PMCID: PMC10957601 DOI: 10.3389/fphar.2024.1337150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 02/27/2024] [Indexed: 03/26/2024] Open
Abstract
Pain is a clinical condition that is currently of great concern and is often caused by tissue or nerve damage or occurs as a concomitant symptom of a variety of diseases such as cancer. Severe pain seriously affects the functional status of the body. However, existing pain management programs are not fully satisfactory. Therefore, there is a need to delve deeper into the pathological mechanisms underlying pain generation and to find new targets for drug therapy. Sphingolipids (SLs), as a major component of the bilayer structure of eukaryotic cell membranes, also have powerful signal transduction functions. Sphingolipids are abundant, and their intracellular metabolism constitutes a huge network. Sphingolipids and their various metabolites play significant roles in cell proliferation, differentiation, apoptosis, etc., and have powerful biological activities. The molecules related to sphingolipid metabolism, mainly the core molecule ceramide and the downstream metabolism molecule sphingosine-1-phosphate (S1P), are involved in the specific mechanisms of neurological disorders as well as the onset and progression of various types of pain, and are closely related to a variety of pain-related diseases. Therefore, sphingolipid metabolism can be the focus of research on pain regulation and provide new drug targets and ideas for pain.
Collapse
Affiliation(s)
- Jianfeng Wang
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Guangda Zheng
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Linfeng Wang
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Linghan Meng
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Juanxia Ren
- Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning Province, China
| | - Lu Shang
- Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning Province, China
| | - Dongtao Li
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | | |
Collapse
|
2
|
Wang X, Feng L, Lu Y, Zhang H. miR-122/PPARβ axis is involved in hypoxic exercise and modulates fatty acid metabolism in skeletal muscle of obese rats. Heliyon 2024; 10:e26572. [PMID: 38434053 PMCID: PMC10906430 DOI: 10.1016/j.heliyon.2024.e26572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 02/11/2024] [Accepted: 02/15/2024] [Indexed: 03/05/2024] Open
Abstract
Hypoxic exercise is an effective intervention for obesity, because it promotes weight loss by regulating fatty acid (FA) metabolism. The regulation of peroxisome proliferator-activated receptor β (PPARβ) by miR-122 may be involved in this process, but the detailed mechanisms are unknown. In order to address this issue, we probed how miR-122 affected the expression of factors associated with FA metabolism in skeletal muscle of obese rats undergoing hypoxic training. By injecting adeno-associated virus 9 containing miR-122 overexpression vector or miR-122 inhibitor into skeletal muscles of rats with a 4-week hypoxic exercise regimen, the miR-122 expression level can be regulated. Body composition and blood lipid levels were analyzed, and PPARβ, carnitine palmitoyltransferase 1b (CPT1b), acetylCoA carboxylase 2 (ACC2), and FA synthase (FAS) mRNA and protein levels were evaluated using quantitative reverse transcription quantitative PCR(RT-qPCR) and Western blot analysis. We found that miR-122 overexpression increased low-density lipoprotein cholesterol (LDL-C) and triglyceride (TG) levels and decreased PPARβ, ACC2, and FAS expression. Conversely, miR-122 inhibition decreased TG level, increased high-density lipoprotein cholesterol (HDL-C) level, and upregulated PPARβ, ACC2, FAS, and CPT1b. These data indicated that the negative regulation of PPARβ by miR-122 promotes FA metabolism by altering the levels of the factors related to FA metabolism in skeletal muscle of obese rat under hypoxic training, thus providing molecular-level insight into the beneficial effects of this intervention.
Collapse
Affiliation(s)
- Xuebing Wang
- College of Physical Education, Guangxi University, Nanning, China
| | - Lianshi Feng
- Exercise Biology Research Center, China Institute of Sport Science, Beijing, China
| | - Yingli Lu
- Exercise Biology Research Center, China Institute of Sport Science, Beijing, China
| | - Haibo Zhang
- Exercise Biology Research Center, China Institute of Sport Science, Beijing, China
| |
Collapse
|
3
|
Wu Q, Xie J, Zhu X, He J. Runt-related transcription factor 3, mediated by DNA-methyltransferase 1, regulated Schwann cell proliferation and myelination during peripheral nerve regeneration via JAK/STAT signaling pathway. Neurosci Res 2023:S0168-0102(23)00008-1. [PMID: 36690210 DOI: 10.1016/j.neures.2023.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 01/15/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023]
Abstract
Schwann cells (SCs) play a crucial role in peripheral nerve injury and regeneration. Recently, RUNX3 was found to be linked with neurological dysfunction. We examined the RUNX3 expression in sciatic nerve stumps with peripheral nerve injury of rats, cyclic adenosine monophosphate (cAMP)-induced SCs. MTT assay was applied to determine the proliferation of SCs. Cell migration and apoptosis were assessed using wound healing assay and flow cytometry. Subsequently, we detected the methylation level of RUNX3 using Methylation-specific PCR assay and verified its regulation by DNMT1. The RUNX3 expressions were increased in sciatic nerve stumps with peripheral nerve injury and cAMP-induced SCs differentiation, which were related to demethylation of its promoter region regulated by DNMT1. RUNX3 knockdown notably suppressed the proliferation and migration, and induced the cell apoptosis of SCs. Silencing of RUNX3 inhibited the cAMP-induced morphological changes of SCs and the increase of myelin-related proteins induced by cAMP in SCs, while RUNX3 overexpression exerted opposite effects. Besides, the overexpression of RUNX3 promoted the activation of JAK/STAT signaling to regulate SCs proliferation and myelination. Meanwhile, DNMT1 overexpression inhibited the expression of RUNX3, and cell proliferation and myelination. In conclusion, RUNX3 mediated by DNMT1 regulated SC proliferation and myelination via JAK/STAT signaling pathway.
Collapse
Affiliation(s)
- Qiufeng Wu
- Department of Neurosurgery, Xianyang Central Hospital, Xianyang, Shaanxi 712000, China
| | - Jiangtao Xie
- Department of Neurosurgery, Xianyang Central Hospital, Xianyang, Shaanxi 712000, China
| | - Xiaoli Zhu
- Department of Neurosurgery, Xianyang Central Hospital, Xianyang, Shaanxi 712000, China
| | - Juan He
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Xianyang, Xianyang, Shaanxi 712000, China.
| |
Collapse
|
4
|
Wang M, Huan Y, Li X, Li J, Lv G. RUNX3 derived hsa_circ_0005752 accelerates the osteogenic differentiation of adipose-derived stem cells via the miR-496/MDM2-p53 pathway. Regen Ther 2021; 18:430-440. [PMID: 34754888 PMCID: PMC8546365 DOI: 10.1016/j.reth.2021.09.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/17/2021] [Accepted: 09/21/2021] [Indexed: 12/18/2022] Open
Abstract
Background Circular RNAs (circRNAs) are non-coding RNAs that play a pivotal role in bone diseases. RUNX3 was an essential transcriptional regulator during osteogenesis. However, it is unknown whether RUNX3 regulates hsa_circ_0005752 during osteogenic differentiation. Methods The levels of hsa_circ_0005752 and RUNX3 were measured by qRT-PCR after osteogenic differentiation of ADSCs. The osteogenic differentiation was analyzed by Alkaline phosphatase (ALP) staining and Alizarin red staining (ARS). qRT-PCR and western blot were used to assess the expressions of osteogenic differentiation-related molecules. RNA pull-down, RIP, and luciferase reporter assays determine the interactions between miR-496 and hsa_circ_0005752 or MDM2 mRNA. CHIP-PCR analyzed the interaction between RUNX3 and LPAR1. Finally, the potential roles of RUNX3 were investigated during osteogenic differentiation with or without hsa_circ_0005752 knockdown. Results Hsa_circ_0005752 and RUNX3 were significantly increased, and miR-496 was remarkably decreased in ADSCs after osteogenic differentiation. Hsa_circ_0005752 could promote osteogenic differentiation, as shown by enhancing ALP and ARS staining intensity. Hsa_circ_0005752 enhanced the expressions of Runx2, ALP, Osx, and OCN. Furthermore, hsa_circ_0005752 directly targeted miR-496, which can directly bind to MDM2. RUNX3 bound to the LPAR1 promoter and enhanced hsa_circ_0005752 expressions. Moreover, the enhanced expression of hsa_circ_0005752 by RUNX3 could promote osteogenic differentiation, whereas knockdown of hsa_circ_0005752 partially antagonized the effects of RUNX3. Conclusion Our study demonstrated that RUNX3 promoted osteogenic differentiation via regulating the hsa_circ_0005752/miR-496/MDM2 axis and thus provided a new therapeutic strategy for osteoporosis.
Collapse
Key Words
- 3′ UTR, 3′ untranslated region
- ADSCs, adipose-derived stem cells
- ALP, alkaline phosphatase
- ARS, Alizarin Red Staining
- Adipose-derived stem cells
- BCA, bicinchoninic acid
- BM-MSCs, Bone Marrow-Mesenchymal Stem Cells
- BMP2, Bone morphogenetic protein 2
- ChIP, chromatin immunoprecipitation
- Circular RNAs
- ECL, enhanced chemiluminescence
- H&E staining, Hematoxylin and Eosin staining
- LPAR1, lysophosphatidic acid receptor 1
- MDM2
- MDM2, murine double minute 2
- OCN, osteocalcin
- OM, osteogenic (differentiation) medium
- Osteogenic differentiation
- Osx, osterix
- PMSF, phenylmethylsulfonyl fluoride
- RIP, RNA immunoprecipitation
- RUNX3
- Runx2, Runt-related transcription factor 2
- Runx3, RUNX Family Transcription Factor 3
- SDS-PAGE, polyacrylamide gel electrophoresis
- UC-MSCs, Umbilical Cord-Mesenchymal Stem Cells
- circRNAs, Circular RNAs
- miRNAs, microRNA
- microRNA
- qRT-PCR, quantitative real-time polymerase chain reaction
Collapse
Affiliation(s)
- Ming Wang
- Department of Spine Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan Province, PR China.,Department of Spine Surgery, The First Affiliated Hospital of University of South China, Hengyang 421001, Hunan Province, PR China
| | - Yifan Huan
- Department of Orthopedics, Financial and Trade Hospital of Hunan Province, Changsha 410001, Hunan Province, PR China
| | - Xiyang Li
- Department of Orthopedics, Financial and Trade Hospital of Hunan Province, Changsha 410001, Hunan Province, PR China
| | - Jing Li
- Department of Spine Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan Province, PR China
| | - Guohua Lv
- Department of Spine Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan Province, PR China
| |
Collapse
|
5
|
Qi Y, Wang W, Song Z, Aji G, Liu XT, Xia P. Role of Sphingosine Kinase in Type 2 Diabetes Mellitus. Front Endocrinol (Lausanne) 2021; 11:627076. [PMID: 33633691 PMCID: PMC7899982 DOI: 10.3389/fendo.2020.627076] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 12/21/2020] [Indexed: 12/15/2022] Open
Abstract
Sphingolipids are a class of essential lipids, functioning as both cell membrane constituents and signaling messengers. In the sphingolipid metabolic network, ceramides serve as the central hub that is hydrolyzed to sphingosine, followed by phosphorylation to sphingosine 1-phosphate (S1P) by sphingosine kinase (SphK). SphK is regarded as a "switch" of the sphingolipid rheostat, as it catalyzes the conversion of ceramide/sphingosine to S1P, which often exhibit opposing biological roles in the cell. Besides, SphK is an important signaling enzyme that has been implicated in the regulation of a wide variety of biological functions. In recent years, an increasing body of evidence has suggested a critical role of SphK in type 2 diabetes mellitus (T2D), although a certain level of controversy remains. Herein, we review recent findings related to SphK in the field of T2D research with a focus on peripheral insulin resistance and pancreatic β-cell failure. It is expected that a comprehensive understanding of the role of SphK and the associated sphingolipids in T2D will help to identify druggable targets for future anti-diabetes therapy.
Collapse
Affiliation(s)
- Yanfei Qi
- Lipid Cell Biology Laboratory, Centenary Institute of Cancer Medicine and Cell Biology, University of Sydney, Sydney, NSW, Australia
| | - Wei Wang
- Department of Endocrinology and Metabolism, Fudan Institute for Metabolic Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ziyu Song
- Department of Endocrinology and Metabolism, Fudan Institute for Metabolic Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Gulibositan Aji
- Department of Endocrinology and Metabolism, Fudan Institute for Metabolic Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xin Tracy Liu
- Lipid Cell Biology Laboratory, Centenary Institute of Cancer Medicine and Cell Biology, University of Sydney, Sydney, NSW, Australia
| | - Pu Xia
- Department of Endocrinology and Metabolism, Fudan Institute for Metabolic Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
6
|
Khan A, Khan A, Khalid S, Shal B, Kang E, Lee H, Laumet G, Seo EK, Khan S. 7β-(3-Ethyl- cis-crotonoyloxy)-1α-(2-methylbutyryloxy)-3,14-dehydro- Z Notonipetranone Attenuates Neuropathic Pain by Suppressing Oxidative Stress, Inflammatory and Pro-Apoptotic Protein Expressions. Molecules 2021; 26:E181. [PMID: 33401491 PMCID: PMC7795484 DOI: 10.3390/molecules26010181] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/19/2020] [Accepted: 12/21/2020] [Indexed: 02/06/2023] Open
Abstract
7β-(3-Ethyl-cis-crotonoyloxy)-1α-(2-methylbutyryloxy)-3,14-dehydro-Z-notonipetranone (ECN), a sesquiterpenoid obtained from a natural source has proved to be effective in minimizing various side effects associated with opioids and nonsteroidal anti-inflammatory drugs. The current study focused on investigating the effects of ECN on neuropathic pain induced by partial sciatic nerve ligation (PSNL) by mainly focusing on oxidative stress, inflammatory and apoptotic proteins expression in mice. ECN (1 and 10 mg/kg, i.p.), was administered once daily for 11 days, starting from the third day after surgery. ECN post-treatment was found to reduce hyperalgesia and allodynia in a dose-dependent manner. ECN remarkably reversed the histopathological abnormalities associated with oxidative stress, apoptosis and inflammation. Furthermore, ECN prevented the suppression of antioxidants (glutathione, glutathione-S-transferase, catalase, superoxide dismutase, NF-E2-related factor-2 (Nrf2), hemeoxygenase-1 and NAD(P)H: quinone oxidoreductase) by PSNL. Moreover, pro-inflammatory cytokines (tumor necrotic factor-alpha, interleukin 1 beta, interleukin 6, cyclooxygenase-2 and inducible nitric oxide synthase) expression was reduced by ECN administration. Treatment with ECN was successful in reducing the caspase-3 level consistent with the observed modulation of pro-apoptotic proteins. Additionally, ECN showed a protective effect on the lipid content of myelin sheath as evident from FTIR spectroscopy which showed the shift of lipid component bands to higher values. Thus, the anti-neuropathic potential of ECN might be due to the inhibition of oxidative stress, inflammatory mediators and pro-apoptotic proteins.
Collapse
Affiliation(s)
- Amna Khan
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan; (A.K.); (A.K.); (S.K.); (B.S.)
| | - Adnan Khan
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan; (A.K.); (A.K.); (S.K.); (B.S.)
| | - Sidra Khalid
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan; (A.K.); (A.K.); (S.K.); (B.S.)
| | - Bushra Shal
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan; (A.K.); (A.K.); (S.K.); (B.S.)
| | - Eunwoo Kang
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea; (E.K.); (H.L.)
| | - Hwaryeong Lee
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea; (E.K.); (H.L.)
| | - Geoffroy Laumet
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA;
| | - Eun Kyoung Seo
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea; (E.K.); (H.L.)
| | - Salman Khan
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan; (A.K.); (A.K.); (S.K.); (B.S.)
| |
Collapse
|
7
|
Chua XY, Ho LTY, Xiang P, Chew WS, Lam BWS, Chen CP, Ong WY, Lai MKP, Herr DR. Preclinical and Clinical Evidence for the Involvement of Sphingosine 1-Phosphate Signaling in the Pathophysiology of Vascular Cognitive Impairment. Neuromolecular Med 2020; 23:47-67. [PMID: 33180310 DOI: 10.1007/s12017-020-08632-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 11/03/2020] [Indexed: 02/07/2023]
Abstract
Sphingosine 1-phosphates (S1Ps) are bioactive lipids that mediate a diverse range of effects through the activation of cognate receptors, S1P1-S1P5. Scrutiny of S1P-regulated pathways over the past three decades has identified important and occasionally counteracting functions in the brain and cerebrovascular system. For example, while S1P1 and S1P3 mediate proinflammatory effects on glial cells and directly promote endothelial cell barrier integrity, S1P2 is anti-inflammatory but disrupts barrier integrity. Cumulatively, there is significant preclinical evidence implicating critical roles for this pathway in regulating processes that drive cerebrovascular disease and vascular dementia, both being part of the continuum of vascular cognitive impairment (VCI). This is supported by clinical studies that have identified correlations between alterations of S1P and cognitive deficits. We review studies which proposed and evaluated potential mechanisms by which such alterations contribute to pathological S1P signaling that leads to VCI-associated chronic neuroinflammation and neurodegeneration. Notably, S1P receptors have divergent but overlapping expression patterns and demonstrate complex interactions. Therefore, the net effect produced by S1P represents the cumulative contributions of S1P receptors acting additively, synergistically, or antagonistically on the neural, vascular, and immune cells of the brain. Ultimately, an optimized therapeutic strategy that targets S1P signaling will have to consider these complex interactions.
Collapse
Affiliation(s)
- Xin Ying Chua
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Leona T Y Ho
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119260, Singapore
| | - Ping Xiang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wee Siong Chew
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Brenda Wan Shing Lam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Christopher P Chen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Kent Ridge, Singapore
| | - Wei-Yi Ong
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119260, Singapore
- Neurobiology Programme, Life Sciences Institute, National University of Singapore, Singapore, 119260, Singapore
| | - Mitchell K P Lai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Memory Aging and Cognition Centre, National University Health System, Kent Ridge, Singapore.
| | - Deron R Herr
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Department of Biology, San Diego State University, San Diego, CA, USA.
- American University of Health Sciences, Long Beach, CA, USA.
| |
Collapse
|
8
|
Squillace S, Spiegel S, Salvemini D. Targeting the Sphingosine-1-Phosphate Axis for Developing Non-narcotic Pain Therapeutics. Trends Pharmacol Sci 2020; 41:851-867. [PMID: 33010954 DOI: 10.1016/j.tips.2020.09.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 09/02/2020] [Accepted: 09/10/2020] [Indexed: 02/07/2023]
Abstract
Chronic pain is a life-altering condition affecting millions of people. Current treatments are inadequate and prolonged therapies come with severe side effects, especially dependence and addiction to opiates. Identification of non-narcotic analgesics is of paramount importance. Preclinical and clinical studies suggest that sphingolipid metabolism alterations contribute to neuropathic pain development. Functional sphingosine-1-phosphate (S1P) receptor 1 (S1PR1) antagonists, such as FTY720/fingolimod, used clinically for non-pain conditions, are emerging as non-narcotic analgesics, supporting the repurposing of fingolimod for chronic pain treatment and energizing drug discovery focused on S1P signaling. Here, we summarize the role of S1P in pain to highlight the potential of targeting the S1P axis towards development of non-narcotic therapeutics, which, in turn, will hopefully help lessen misuse of opioid pain medications and address the ongoing opioid epidemic.
Collapse
Affiliation(s)
- Silvia Squillace
- Department of Pharmacology and Physiology and the Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
| | - Daniela Salvemini
- Department of Pharmacology and Physiology and the Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, MO 63104, USA.
| |
Collapse
|
9
|
Wang X, Lu Y, Zhu L, Zhang H, Feng L. Inhibition of miR-27b Regulates Lipid Metabolism in Skeletal Muscle of Obese Rats During Hypoxic Exercise by Increasing PPARγ Expression. Front Physiol 2020; 11:1090. [PMID: 32982800 PMCID: PMC7489097 DOI: 10.3389/fphys.2020.01090] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 08/07/2020] [Indexed: 12/12/2022] Open
Abstract
Hypoxic exercise may represent a novel therapeutic strategy to reduce and prevent obesity through the regulation of lipid metabolism. During hypoxic exercise, the targeting of peroxisome proliferator-activated receptor gamma (PPARγ) by miR-27b has been proposed to be one of the mechanisms involved in the modulation of lipid metabolism. We have previously shown that miR-27b can repress PPARγ and lipid metabolism-associated factors, thereby affecting lipid metabolism during hypoxic exercise in a rat model of obesity. In the current study, we aimed to confirm the role of miR-27b in the regulation of lipid metabolism. First, miR-27b expression was either upregulated or downregulated through the injection of adeno-associated virus (AAV) 9 containing a miR-27b expression cassette or miR-27b-3p inhibitor, respectively, into the right gastrocnemius muscle of obese rats. The rats were then subjected to a 4-week program of hypoxic exercise, and a series of parameters related to lipid metabolism were systematically evaluated, including body composition, blood lipid levels, miR-27b RNA levels, and mRNA and protein levels of PPARγ and those of its downstream lipid metabolism-associated factors. No significant differences were found in body composition between rats expressing different levels of miR-27b. However, regarding blood lipids, miR-27b overexpression led to increased concentrations of triglycerides (TG), low-density lipoprotein cholesterol (LDL-C), and free fatty acids (FFAs), while inhibition of miR-27b decreased the total cholesterol (TC) level and increased that of high-density lipoprotein cholesterol (HDL-C). At the mRNA level, miR-27b overexpression downregulated the expression of Pparγ, but upregulated that of lipid metabolism-associated factors such as heart-type fatty acid-binding protein (H-FABP), fatty acid transport protein 1 (FATP1), adipose triglyceride lipase (ATGL), and lipoprotein lipase (LPL), whereas miR-27b inhibition elicited the opposite effect; however, inhibition of miR-27b led to elevated cholesterol 7 alpha-hydroxylase (CYP7A1) and fatty acid translocase 36 (CD36) levels. Similarly, at the protein level, miR-27b overexpression promoted a decrease in the concentration of PPARγ, whereas miR-27b inhibition led to an increase in PPARγ levels, as well as those of CYP7A1, CD36, ATGL, and LPL. Overall, our results indicated that hypoxic exercise regulates lipid metabolism via the miR-27b/PPARγ pathway and modulates ATGL and LPL expression through inducing their post-transcriptional modifications.
Collapse
Affiliation(s)
- Xuebing Wang
- School of Kinesiology, Shanghai University of Sport, Shanghai, China.,College of Physical Education, Guangxi University, Nanning, China
| | - Yingli Lu
- Exercise Biology Research Center, China Institute of Sport Science, Beijing, China
| | - Lei Zhu
- School of Sports Science, Qufu Normal University, Qufu, China
| | - Haibo Zhang
- Exercise Biology Research Center, China Institute of Sport Science, Beijing, China
| | - Lianshi Feng
- Exercise Biology Research Center, China Institute of Sport Science, Beijing, China
| |
Collapse
|
10
|
Jiang P, Jiang Q, Yan Y, Hou Z, Luo D. Propofol ameliorates neuropathic pain and neuroinflammation through PPAR γ up-regulation to block Wnt/β-catenin pathway. Neurol Res 2020; 43:71-77. [PMID: 32985377 DOI: 10.1080/01616412.2020.1823107] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE As an intravenous anesthetic, propofol has been exhibited to provide excellent clinical analgesia. Whether propofol has amelioration property for NP and neuroinflammation remains unexplored. The present study was arranged to probe the role of propofol in the mitigation of NP and neuroinflammation in rats and underlying mechanisms. METHODS Rats were randomly classified into the following groups: Model, Sham, Control, Propofol, GW9662, and Saline groups. The radiant heat stimulation was used to measure paw withdrawal latency (PWL), and mechanical stimulation was employed to detect paw withdrawal threshold (PWT). Subsequently, the expression of GFAP was assessed by immunofluorescence to reflect the activation of astrocyte. qRT-PCR and Western blot were utilized for the performance of mRNA and protein expression levels of PPAR γ as well as inflammation factors (TNF-α, IL-1β, and IL-6). RESULTS Pentobarbital sodium anesthesia significantly shortened the PWL and PWT, suppressed PPAR γ expression in rats in addition to elevating astrocyte activation and inflammation response. Propofol treatment attenuated the NP of rats as evidenced by restrained astrocyte activation level and inflammation factor levels. Rats treated with propofol had markedly heightened PPAR γ expression. PPAR γ exposure ameliorated NP and inflammation degree, which demonstrated by elevated astrocyte activation and inflammation levels as well as suppressed PWL and PWT in rats injected with PPAR γ inhibitor. Besides, PPAR γ decreased the expression level of β-catenin. CONCLUSION Propofol ameliorates NP and neuroinflammation of rats by up-regulating PPAR γ expression to block the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Peng Jiang
- Department of Anesthesiology, Huizhou Municipal Central Hospital , Huizhou, Guangdong, P.R. China
| | - Qun Jiang
- Department of Anesthesiology, Traditional Chinese Medicine Hospital of Guangdong Province , Guangzhou, Guangdong, P.R. China
| | - Yan Yan
- Department of Anesthesiology, Huizhou Municipal Central Hospital , Huizhou, Guangdong, P.R. China
| | - Zhiqi Hou
- Department of Anesthesiology, Huizhou Municipal Central Hospital , Huizhou, Guangdong, P.R. China
| | - Dexing Luo
- Department of Anesthesiology, Huizhou Municipal Central Hospital , Huizhou, Guangdong, P.R. China
| |
Collapse
|
11
|
Jiang X, Wang S, Chen H. A Novel Fabrication of Dose-Dependent Injectable Curcumin Biocomposite Hydrogel System Anesthetic Delivery Method for Care and Management of Musculoskeletal Pain. Dose Response 2020; 18:1559325820929555. [PMID: 32782446 PMCID: PMC7385839 DOI: 10.1177/1559325820929555] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 04/28/2020] [Accepted: 04/30/2020] [Indexed: 12/30/2022] Open
Abstract
Chronic musculoskeletal pain has biological, psychological, and social components. In this article, we have demonstrated the easily injectable nanocomposite carrier for the treatment of chronic musculoskeletal pain. Briefly, the curcumin (Cur) loaded with lipid nanocapsules (LNCs; Cur@LNCs) using the phase invasion method. The synthesized Cur@LNCs were characterized by using scanning electron microscopy, transmittance electron microscopy, and the size of the fabricated nanoparticles confirmed by dynamic light scattering analysis. The synthesized Cur@LNC injectable hydrogel shows excellent results in vivo in the rat model. We have examined the efficiency of the chronic constriction injury in the rat model and induced the pain using thermal paw withdrawal latency. The injectable hydrogels Cur@LNCs display a remarkable reduction in pain 7 days post administrations compared to the untreated group animals. This work could establish the preclinical candidate of the neuropathic pain response in the future.
Collapse
Affiliation(s)
- Xuehong Jiang
- Department of General Medicine, First People’s Hospital of Wenling, Wenling, China
- Department of Spinal Surgery, First People’s Hospital of Wenling, Wenling, China
| | - Shuaishuai Wang
- Department of General Medicine, First People’s Hospital of Wenling, Wenling, China
- Department of Spinal Surgery, First People’s Hospital of Wenling, Wenling, China
| | - Hui Chen
- Department of General Medicine, First People’s Hospital of Wenling, Wenling, China
- Department of Spinal Surgery, First People’s Hospital of Wenling, Wenling, China
| |
Collapse
|
12
|
Langeslag M, Kress M. The ceramide-S1P pathway as a druggable target to alleviate peripheral neuropathic pain. Expert Opin Ther Targets 2020; 24:869-884. [PMID: 32589067 DOI: 10.1080/14728222.2020.1787989] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Introduction: Neuropathic pain disorders are diverse, and the currently available therapies are ineffective in the majority of cases. Therefore, there is a major need for gaining novel mechanistic insights and developing new treatment strategies for neuropathic pain. Areas covered: We performed an in-depth literature search on the molecular mechanisms and systemic importance of the ceramide-to-S1P rheostat regulating neuron function and neuroimmune interactions in the development of neuropathic pain. Expert opinion: The S1P receptor modulator FTY720 (fingolimod, Gilenya®), LPA receptor antagonists and several mechanistically related compounds in clinical development raise great expectations for treating neuropathic pain disorders. Research on S1P receptors, S1P receptor modulators or SPHK inhibitors with distinct selectivity, pharmacokinetics and safety must provide more mechanistic insight into whether they may qualify as useful treatment options for neuropathic pain disorders. The functional relevance of genetic variations within the ceramide-to-S1P rheostat should be explored for an enhanced understanding of neuropathic pain pathogenesis. The ceramide-to-S1P rheostat is emerging as a critically important regulator hub of neuroimmune interactions along the pain pathway, and improved mechanistic insight is required to develop more precise and effective drug treatment options for patients suffering from neuropathic pain disorders.
Collapse
Affiliation(s)
- Michiel Langeslag
- Institute of Physiology, DPMP, Medical University Innsbruck , Austria
| | - Michaela Kress
- Institute of Physiology, DPMP, Medical University Innsbruck , Austria
| |
Collapse
|
13
|
Sphingosine 1-Phosphate Receptor 2 Induces Otoprotective Responses to Cisplatin Treatment. Cancers (Basel) 2020; 12:cancers12010211. [PMID: 31952197 PMCID: PMC7016659 DOI: 10.3390/cancers12010211] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 11/08/2019] [Accepted: 01/09/2020] [Indexed: 12/19/2022] Open
Abstract
Ototoxicity is a major adverse effect of platinum-based chemotherapeutics and currently, there remains a lack of United States Food and Drug Administration-approved therapies to prevent or treat this problem. In our study, we examined the role of the sphingosine 1-phosphate receptor 2 (S1P2) in attenuating cisplatin-induced ototoxicity in several different animal models and cell lines. We found that ototoxicity in S1P2 knockout mice is dependent on reactive oxygen species (ROS) production and that S1P2 receptor activation with a specific agonist, CYM-5478, significantly attenuates cisplatin-induced defects, including hair cell degeneration in zebrafish and prolonged auditory brainstem response latency in rats. We also evaluated the cytoprotective effect of CYM-5478 across different cell lines and showed that CYM-5478 protects neural-derived cell lines but not breast cancer cells against cisplatin toxicity. We show that this selective protection of CYM-5478 is due to its differential effects on key regulators of apoptosis between neural cells and breast cancer cells. Overall, our study suggests that targeting the S1P2 receptor represents a promising therapeutic approach for the treatment of cisplatin-induced ototoxicity in cancer patients.
Collapse
|
14
|
Activation of sphingosine 1-phosphate receptor 2 attenuates chemotherapy-induced neuropathy. J Biol Chem 2020. [DOI: 10.1016/s0021-9258(17)49922-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
15
|
Wang W, Xiang P, Chew WS, Torta F, Bandla A, Lopez V, Seow WL, Lam BWS, Chang JK, Wong P, Chayaburakul K, Ong WY, Wenk MR, Sundar R, Herr DR. Activation of sphingosine 1-phosphate receptor 2 attenuates chemotherapy-induced neuropathy. J Biol Chem 2019; 295:1143-1152. [PMID: 31882542 DOI: 10.1074/jbc.ra119.011699] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/07/2019] [Indexed: 12/11/2022] Open
Abstract
Platinum-based therapeutics are used to manage many forms of cancer, but frequently result in peripheral neuropathy. Currently, the only option available to attenuate chemotherapy-induced neuropathy is to limit or discontinue this treatment. Sphingosine 1-phosphate (S1P) is a lipid-based signaling molecule involved in neuroinflammatory processes by interacting with its five cognate receptors: S1P1-5 In this study, using a combination of drug pharmacodynamic analysis in human study participants, disease modeling in rodents, and cell-based assays, we examined whether S1P signaling may represent a potential target in the treatment of chemotherapy-induced neuropathy. To this end, we first investigated the effects of platinum-based drugs on plasma S1P levels in human cancer patients. Our analysis revealed that oxaliplatin treatment specifically increases one S1P species, d16:1 S1P, in these patients. Although d16:1 S1P is an S1P2 agonist, it has lower potency than the most abundant S1P species (d18:1 S1P). Therefore, as d16:1 S1P concentration increases, it is likely to disproportionately activate proinflammatory S1P1 signaling, shifting the balance away from S1P2 We further show that a selective S1P2 agonist, CYM-5478, reduces allodynia in a rat model of cisplatin-induced neuropathy and attenuates the associated inflammatory processes in the dorsal root ganglia, likely by activating stress-response proteins, including ATF3 and HO-1. Cumulatively, the findings of our study suggest that the development of a specific S1P2 agonist may represent a promising therapeutic approach for the management of chemotherapy-induced neuropathy.
Collapse
Affiliation(s)
- Wei Wang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228
| | - Ping Xiang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228
| | - Wee Siong Chew
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228
| | - Federico Torta
- Singapore Lipidomics Incubator (SLING), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228
| | - Aishwarya Bandla
- The N.1 Institute for Health, National University of Singapore, Singapore 119077
| | - Violeta Lopez
- Alice Lee Centre for Nursing Studies, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228
| | - Wei Lun Seow
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228
| | - Brenda Wan Shing Lam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228
| | - Jing Kai Chang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228
| | - Peiyan Wong
- Neuroscience Phenotyping Core, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228
| | | | - Wei-Yi Ong
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228.,Neurobiology and Ageing Research Programme, National University of Singapore, Singapore 119077
| | - Markus R Wenk
- Singapore Lipidomics Incubator (SLING), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228
| | - Raghav Sundar
- The N.1 Institute for Health, National University of Singapore, Singapore 119077 .,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228.,Department of Haematology-Oncology, National University Health System, Singapore 119074
| | - Deron R Herr
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228 .,Department of Biology, San Diego State University, San Diego, California 92182
| |
Collapse
|