1
|
Zhao M, Bian R, Xu X, Zhang J, Zhang L, Zheng Y. Sphingolipid Metabolism and Signalling Pathways in Heart Failure: From Molecular Mechanism to Therapeutic Potential. J Inflamm Res 2025; 18:5477-5498. [PMID: 40291458 PMCID: PMC12034266 DOI: 10.2147/jir.s515757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Accepted: 04/16/2025] [Indexed: 04/30/2025] Open
Abstract
Sphingolipids are essential components of cell membranes and lipoproteins. They are synthesized de novo in the endoplasmic reticulum and subsequently undergo various enzymatic modifications in different organelles, giving rise to a diverse range of biologically active compounds. These molecules play a critical role in regulating cell growth, senescence, migration, apoptosis, and signaling. In recent years, the sphingolipid metabolic pathway has been recognized as a key factor in heart failure (HF) pathophysiology. Abnormal levels of sphingolipid metabolites, such as ceramide (Cer) and sphingomyelin (SM), contribute to oxidative stress and inflammatory responses, ultimately promoting cardiomyocyte apoptosis. Conversely, sphingosine-1-phosphate (S1P) and ceramide-1-phosphate (C1P) regulate vascular function and influence cardiac remodeling. Additionally, enzymes such as diacylglycerol acyltransferase 1 (DGAT1) and sphingosine-1-phosphate lyase 1 (SGPL1) modulate cardiac lipid metabolism. Given their role in HF progression, monitoring sphingolipid alterations offers potential as valuable biomarkers for assessing disease severity, prognosis, and diagnosis. Given the complexity of sphingolipid metabolism and its involvement in diverse regulatory biological processes, a comprehensive understanding of its roles at both the cellular and organismal levels in physiopathology remains incomplete. Therefore, this review aims to explore the physiological functions, regulatory mechanisms, and therapeutic potential of sphingolipid metabolism. It will summarize the specific molecular mechanisms driving key pathological processes in HF, including ventricular remodeling, myocardial fibrosis, vascular dysfunction, and metabolic disorders. Finally, the review will highlight targeted sphingolipid metabolites as potential therapeutic strategies, offering new insights into HF diagnosis and treatment, with the goal of advancing adjunctive clinical therapies.
Collapse
Affiliation(s)
- Meng Zhao
- The First Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, Henan Province, People’s Republic of China
- Department of Cardiology, Zhengzhou Hospital of Traditional Chinese Medicine, Zhengzhou, Henan Province, People’s Republic of China
- Joint Formula and Syndrome Research Laboratory of Guangzhou University of Chinese Medicine & Zhengzhou Hospital of Chinese Medicine, Zhengzhou, Henan Province, People’s Republic of China
| | - Rutao Bian
- Department of Cardiology, Zhengzhou Hospital of Traditional Chinese Medicine, Zhengzhou, Henan Province, People’s Republic of China
| | - Xuegong Xu
- Department of Cardiology, Zhengzhou Hospital of Traditional Chinese Medicine, Zhengzhou, Henan Province, People’s Republic of China
| | - Junpeng Zhang
- Department of Cardiology, Zhengzhou Hospital of Traditional Chinese Medicine, Zhengzhou, Henan Province, People’s Republic of China
| | - Li Zhang
- Department of Cardiology, Zhengzhou Hospital of Traditional Chinese Medicine, Zhengzhou, Henan Province, People’s Republic of China
| | - Yi Zheng
- Department of Cardiology, Zhengzhou Hospital of Traditional Chinese Medicine, Zhengzhou, Henan Province, People’s Republic of China
| |
Collapse
|
2
|
Zhu Z, Tu B, Peng C, Xu X, Lu P, Ning R. Integrated bioinformatics and clinical data identify three novel biomarkers for osteoarthritis diagnosis and synovial immune. Sci Rep 2025; 15:10987. [PMID: 40164659 PMCID: PMC11958655 DOI: 10.1038/s41598-025-95837-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 03/24/2025] [Indexed: 04/02/2025] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease that can be aggravated by synovitis and synovial immune disorders (SID). However, the role of synovial SID-related genes in OA synovium remains poorly understood. OA synovial and peripheral blood datasets were obtained from the GEO database ( https://www.ncbi.nlm.nih.gov/ ). Immune-related genes ( https://reactome.org/ ) showing differential expression in peripheral blood were identified as immune disorder genes. Subsequently, differentially expressed immune disorder genes in OA synovium were further identified as SID genes. The Venn diagram, random forest, SVM-RFE algorithm, and multivariate analysis were employed to determine SID-related hub genes in OA synovium. Using the identified hub genes, we constructed and validated a diagnostic model for predicting OA occurrence. The correlation between hub gene expression and immune-related modules was explored using CIBERSORT and MCP-counter analyses. We identified three SID-related hub genes (ACAT1, SPHK1, and ACACB) in OA synovium. The diagnostic model incorporating these hub genes demonstrated reliable predictive accuracy (AUC = 0.939). Through qPCR analysis, we quantitated the expression levels of the hub genes and confirmed that three hub genes could serve as novel biomarkers for OA patients (AUC = 0.960). Furthermore, we observed a significant correlation between the expression of these hub genes and immune cell infiltration, as well as inflammatory cytokine levels in OA synovium. Our findings suggest that three SID-related hub genes have the potential to serve as diagnostic biomarkers for OA patients. These genes are associated with immune disorder and contribute to immune alterations within the OA synovium.
Collapse
Affiliation(s)
- Zheng Zhu
- Department of Orthopedics, Hefei First People's Hospital, Anhui Medical University, 390 Huaihe Road, Hefei, 230061, Anhui, China
| | - Bizhi Tu
- Department of Orthopedics, Hefei First People's Hospital, Anhui Medical University, 390 Huaihe Road, Hefei, 230061, Anhui, China
| | - Cheng Peng
- Department of Orthopedics, Hefei First People's Hospital, Anhui Medical University, 390 Huaihe Road, Hefei, 230061, Anhui, China
| | - Xun Xu
- Department of Orthopedics, Hefei First People's Hospital, Anhui Medical University, 390 Huaihe Road, Hefei, 230061, Anhui, China
| | - Peizhi Lu
- Department of Orthopedics, Hefei First People's Hospital, Anhui Medical University, 390 Huaihe Road, Hefei, 230061, Anhui, China
| | - Rende Ning
- Department of Orthopedics, Hefei First People's Hospital, Anhui Medical University, 390 Huaihe Road, Hefei, 230061, Anhui, China.
| |
Collapse
|
3
|
Ge T, Ning B, Wu Y, Chen X, Qi H, Wang H, Zhao M. MicroRNA-specific therapeutic targets and biomarkers of apoptosis following myocardial ischemia-reperfusion injury. Mol Cell Biochem 2024; 479:2499-2521. [PMID: 37878166 DOI: 10.1007/s11010-023-04876-z] [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: 07/12/2023] [Accepted: 10/05/2023] [Indexed: 10/26/2023]
Abstract
MicroRNAs are single-stranded non-coding RNAs that participate in post-transcriptional regulation of gene expression, it is involved in the regulation of apoptosis after myocardial ischemia-reperfusion injury. For example, the alteration of mitochondrial structure is facilitated by MicroRNA-1 through the regulation of apoptosis-related proteins, such as Bax and Bcl-2, thereby mitigating cardiomyocyte apoptosis. MicroRNA-21 not only modulates the expression of NF-κB to suppress inflammatory signals but also activates the PI3K/AKT pathway to mitigate ischemia-reperfusion injury. Overexpression of MicroRNA-133 attenuates reactive oxygen species (ROS) production and suppressed the oxidative stress response, thereby mitigating cellular apoptosis. MicroRNA-139 modulates the extrinsic death signal of Fas, while MicroRNA-145 regulates endoplasmic reticulum calcium overload, both of which exert regulatory effects on cardiomyocyte apoptosis. Therefore, the article categorizes the molecular mechanisms based on the three classical pathways and multiple signaling pathways of apoptosis. It summarizes the targets and pathways of MicroRNA therapy for ischemia-reperfusion injury and analyzes future research directions.
Collapse
Affiliation(s)
- Teng Ge
- School of Graduate, Shaanxi University of Chinese Medicine, No. 1 Middle Section of Shiji Avenue, Xianyang, 712046, China
| | - Bo Ning
- School of Graduate, Shaanxi University of Chinese Medicine, No. 1 Middle Section of Shiji Avenue, Xianyang, 712046, China
| | - Yongqing Wu
- School of Graduate, Shaanxi University of Chinese Medicine, No. 1 Middle Section of Shiji Avenue, Xianyang, 712046, China
| | - Xiaolin Chen
- School of Pharmacy, Shaanxi University of Chinese Medicine, No. 1 Middle Section of Shiji Avenue, Xianyang, 712046, China
| | - Hongfei Qi
- Shaanxi Key Laboratory of Integrated Traditional and Western Medicine for Prevention and Treatment of Cardiovascular Diseases, Institute of Integrative Medicine, Shaanxi University of Chinese Medicine, No. 1 Middle Section of Shiji Avenue, Xianyang, 712046, China
| | - Haifang Wang
- Shaanxi Key Laboratory of Integrated Traditional and Western Medicine for Prevention and Treatment of Cardiovascular Diseases, Institute of Integrative Medicine, Shaanxi University of Chinese Medicine, No. 1 Middle Section of Shiji Avenue, Xianyang, 712046, China
| | - Mingjun Zhao
- Department of Cardiology, Affiliated Hospital of Shaanxi University of Chinese Medicine, Deputy 2, Weiyang West Road, Weicheng District, Xianyang, 712000, China.
| |
Collapse
|
4
|
Jin L, Zhu J, Yao L, Shen G, Xue BX, Tao W. Targeting SphK1/2 by SKI-178 inhibits prostate cancer cell growth. Cell Death Dis 2023; 14:537. [PMID: 37604912 PMCID: PMC10442381 DOI: 10.1038/s41419-023-06023-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 07/13/2023] [Accepted: 07/27/2023] [Indexed: 08/23/2023]
Abstract
Sphingosine kinases (SphK), including SphK1 and SphK2, are important enzymes promoting progression of prostate cancer. SKI-178 is a novel and highly potent SphK1/2 dual inhibitor. We here tested the potential anti-prostate cancer cell activity of SKI-178. Bioinformatics analyses and results from local tissues demonstrated that that both SphK1 and SphK2 are upregulated in human prostate cancer tissues. Ectopic overexpression of SphK1 and SphK2, by lentiviral constructs, promoted primary prostate cancer cell proliferation and migration. In primary human prostate cancer cells and immortalized cell lines, SKI-178 potently inhibited cell viability, proliferation, cell cycle progression and cell migration, causing robust cell death and apoptosis. SKI-178 impaired mitochondrial functions, causing mitochondrial depolarization, reactive oxygen species production and ATP depletion.SKI-178 potently inhibited SphK activity and induced ceramide production, without affecting SphK1/2 expression in prostate cancer cells. Further, SKI-178 inhibited Akt-mTOR activation and induced JNK activation in prostate cancer cells. Contrarily, a constitutively-active Akt1 construct or the pharmacological JNK inhibitors attenuated SKI-178-induced cytotoxicity in prostate cancer cells. In vivo, daily intraperitoneal injection of a single dose of SKI-178 potently inhibited PC-3 xenograft growth in nude mice. SphK inhibition, ceramide production, ATP depletion and lipid peroxidation as well as Akt-mTOR inactivation and JNK activation were detected in PC-3 xenograft tissues with SKI-178 administration. Together, targeting SphK1/2 by SKI-178 potently inhibited prostate cancer cell growth in vitro and in vivo.
Collapse
Affiliation(s)
- Lu Jin
- Department of Urology, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jin Zhu
- Department of Urology, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Linya Yao
- Department of Urology, Kunshan Hospital of Traditional Chinese Medicine Affiliated to Yangzhou University, Kunshan, China
| | - Gang Shen
- Department of Urology, DUSHU Lake Hospital Affiliated to Soochow University, Suzhou, China.
| | - Bo-Xin Xue
- Department of Urology, the Second Affiliated Hospital of Soochow University, Suzhou, China.
| | - Wei Tao
- Department of Urology, the Second Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
5
|
Ya'ar Bar S, Pintel N, Abd Alghne H, Khattib H, Avni D. The therapeutic potential of sphingolipids for cardiovascular diseases. Front Cardiovasc Med 2023; 10:1224743. [PMID: 37608809 PMCID: PMC10440740 DOI: 10.3389/fcvm.2023.1224743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/17/2023] [Indexed: 08/24/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of morbidity and mortality worldwide and Inflammation plays a critical role in the development of CVD. Despite considerable progress in understanding the underlying mechanisms and various treatment options available, significant gaps in therapy necessitate the identification of novel therapeutic targets. Sphingolipids are a family of lipids that have gained attention in recent years as important players in CVDs and the inflammatory processes that underlie their development. As preclinical studies have shown that targeting sphingolipids can modulate inflammation and ameliorate CVDs, targeting sphingolipids has emerged as a promising therapeutic strategy. This review discusses the current understanding of sphingolipids' involvement in inflammation and cardiovascular diseases, the existing therapeutic approaches and gaps in therapy, and explores the potential of sphingolipids-based drugs as a future avenue for CVD treatment.
Collapse
Affiliation(s)
- Sapir Ya'ar Bar
- Department of Natural Compound, Nutrition, and Health, MIGAL, Kiryat Shmona, Israel
| | - Noam Pintel
- Department of Natural Compound, Nutrition, and Health, MIGAL, Kiryat Shmona, Israel
| | - Hesen Abd Alghne
- Department of Natural Compound, Nutrition, and Health, MIGAL, Kiryat Shmona, Israel
- Tel-Hai College Department of Biotechnology, Kiryat Shmona, Israel
| | - Hamdan Khattib
- Department of Natural Compound, Nutrition, and Health, MIGAL, Kiryat Shmona, Israel
- Department of Gastroenterology and Hepatology, Tel Aviv University Sackler Faculty of Medicine, Tel Aviv, Israel
| | - Dorit Avni
- Department of Natural Compound, Nutrition, and Health, MIGAL, Kiryat Shmona, Israel
- Tel-Hai College Department of Biotechnology, Kiryat Shmona, Israel
| |
Collapse
|
6
|
Cross-Regulation of the Cellular Redox System, Oxygen, and Sphingolipid Signalling. Metabolites 2023; 13:metabo13030426. [PMID: 36984866 PMCID: PMC10054022 DOI: 10.3390/metabo13030426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023] Open
Abstract
Redox-active mediators are now appreciated as powerful molecules to regulate cellular dynamics such as viability, proliferation, migration, cell contraction, and relaxation, as well as gene expression under physiological and pathophysiological conditions. These molecules include the various reactive oxygen species (ROS), and the gasotransmitters nitric oxide (NO∙), carbon monoxide (CO), and hydrogen sulfide (H2S). For each of these molecules, direct targets have been identified which transmit the signal from the cellular redox state to a cellular response. Besides these redox mediators, various sphingolipid species have turned out as highly bioactive with strong signalling potential. Recent data suggest that there is a cross-regulation existing between the redox mediators and sphingolipid molecules that have a fundamental impact on a cell’s fate and organ function. This review will summarize the effects of the different redox-active mediators on sphingolipid signalling and metabolism, and the impact of this cross-talk on pathophysiological processes. The relevance of therapeutic approaches will be highlighted.
Collapse
|
7
|
Borodzicz-Jażdżyk S, Jażdżyk P, Łysik W, Cudnoch-Jȩdrzejewska A, Czarzasta K. Sphingolipid metabolism and signaling in cardiovascular diseases. Front Cardiovasc Med 2022; 9:915961. [PMID: 36119733 PMCID: PMC9471951 DOI: 10.3389/fcvm.2022.915961] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 08/05/2022] [Indexed: 01/10/2023] Open
Abstract
Sphingolipids are a structural component of the cell membrane, derived from sphingosine, an amino alcohol. Its sphingoid base undergoes various types of enzymatic transformations that lead to the formation of biologically active compounds, which play a crucial role in the essential pathways of cellular signaling, proliferation, maturation, and death. The constantly growing number of experimental and clinical studies emphasizes the pivotal role of sphingolipids in the pathophysiology of cardiovascular diseases, including, in particular, ischemic heart disease, hypertension, heart failure, and stroke. It has also been proven that altering the sphingolipid metabolism has cardioprotective properties in cardiac pathologies, including myocardial infarction. Recent studies suggest that selected sphingolipids may serve as valuable biomarkers useful in the prognosis of cardiovascular disorders in clinical practice. This review aims to provide an overview of the current knowledge of sphingolipid metabolism and signaling in cardiovascular diseases.
Collapse
Affiliation(s)
- Sonia Borodzicz-Jażdżyk
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Piotr Jażdżyk
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
- Second Department of Psychiatry, Institute of Psychiatry and Neurology in Warsaw, Warsaw, Poland
| | - Wojciech Łysik
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Agnieszka Cudnoch-Jȩdrzejewska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Katarzyna Czarzasta
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
8
|
Targeting sphingosine kinase 1/2 by a novel dual inhibitor SKI-349 suppresses non-small cell lung cancer cell growth. Cell Death Dis 2022; 13:602. [PMID: 35831279 PMCID: PMC9279331 DOI: 10.1038/s41419-022-05049-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/16/2022] [Accepted: 06/27/2022] [Indexed: 01/21/2023]
Abstract
Sphingosine kinase 1 (SphK1) and sphingosine kinase (SphK2) are both important therapeutic targets of non-small cell lung cancer (NSCLC). SKI-349 is a novel, highly efficient and small molecular SphK1/2 dual inhibitor. Here in primary human NSCLC cells and immortalized cell lines, SKI-349 potently inhibited cell proliferation, cell cycle progression, migration and viability. The dual inhibitor induced mitochondrial depolarization and apoptosis activation in NSCLC cells, but it was non-cytotoxic to human lung epithelial cells. SKI-349 inhibited SphK activity and induced ceramide accumulation in primary NSCLC cells, without affecting SphK1/2 expression. SKI-349-induced NSCLC cell death was attenuated by sphingosine-1-phosphate and by the SphK activator K6PC-5, but was potentiated by the short-chain ceramide C6. Moreover, SKI-349 induced Akt-mTOR inactivation, JNK activation, and oxidative injury in primary NSCLC cells. In addition, SKI-349 decreased bromodomain-containing protein 4 (BRD4) expression and downregulated BRD4-dependent genes (Myc, cyclin D1 and Klf4) in primary NSCLC cells. At last, SKI-349 (10 mg/kg) administration inhibited NSCLC xenograft growth in nude mice. Akt-mTOR inhibition, JNK activation, oxidative injury and BRD4 downregulation were detected in SKI-349-treated NSCLC xenograft tissues. Taken together, targeting SphK1/2 by SKI-349 potently inhibits NSCLC cell growth in vitro and in vivo.
Collapse
|
9
|
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: 3.0] [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
|
10
|
Ueda N. A Rheostat of Ceramide and Sphingosine-1-Phosphate as a Determinant of Oxidative Stress-Mediated Kidney Injury. Int J Mol Sci 2022; 23:ijms23074010. [PMID: 35409370 PMCID: PMC9000186 DOI: 10.3390/ijms23074010] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/29/2022] [Accepted: 03/29/2022] [Indexed: 02/06/2023] Open
Abstract
Reactive oxygen species (ROS) modulate sphingolipid metabolism, including enzymes that generate ceramide and sphingosine-1-phosphate (S1P), and a ROS-antioxidant rheostat determines the metabolism of ceramide-S1P. ROS induce ceramide production by activating ceramide-producing enzymes, leading to apoptosis, while they inhibit S1P production, which promotes survival by suppressing sphingosine kinases (SphKs). A ceramide-S1P rheostat regulates ROS-induced mitochondrial dysfunction, apoptotic/anti-apoptotic Bcl-2 family proteins and signaling pathways, leading to apoptosis, survival, cell proliferation, inflammation and fibrosis in the kidney. Ceramide inhibits the mitochondrial respiration chain and induces ceramide channel formation and the closure of voltage-dependent anion channels, leading to mitochondrial dysfunction, altered Bcl-2 family protein expression, ROS generation and disturbed calcium homeostasis. This activates ceramide-induced signaling pathways, leading to apoptosis. These events are mitigated by S1P/S1P receptors (S1PRs) that restore mitochondrial function and activate signaling pathways. SphK1 promotes survival and cell proliferation and inhibits inflammation, while SphK2 has the opposite effect. However, both SphK1 and SphK2 promote fibrosis. Thus, a ceramide-SphKs/S1P rheostat modulates oxidant-induced kidney injury by affecting mitochondrial function, ROS production, Bcl-2 family proteins, calcium homeostasis and their downstream signaling pathways. This review will summarize the current evidence for a role of interaction between ROS-antioxidants and ceramide-SphKs/S1P and of a ceramide-SphKs/S1P rheostat in the regulation of oxidative stress-mediated kidney diseases.
Collapse
Affiliation(s)
- Norishi Ueda
- Department of Pediatrics, Public Central Hospital of Matto Ishikawa, 3-8 Kuramitsu, Hakusan 924-8588, Japan
| |
Collapse
|
11
|
Sun X, Shan HJ, Yin G, Zhang XY, Huang YM, Li HJ. The anti-osteosarcoma cell activity by the sphingosine kinase 1 inhibitor SKI-V. Cell Death Dis 2022; 8:48. [PMID: 35115496 PMCID: PMC8814198 DOI: 10.1038/s41420-022-00838-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 01/03/2022] [Accepted: 01/14/2022] [Indexed: 01/04/2023]
Abstract
Sphingosine kinase 1 (SphK1) expression and activity are elevated in human osteosarcoma (OS) and is a promising target of therapy. SKI-V is a non-competitive and highly-efficient non-lipid SphK1 inhibitor. The potential anti-OS cell activity by the SphK1 inhibitor was studied here. In primary OS cells and immortalized cell lines, SKI-V robustly suppressed cell survival, growth and proliferation as well as cell mobility, and inducing profound OS cell death and apoptosis. The SphK1 inhibitor was however non-cytotoxic nor pro-apoptotic in human osteoblasts. SKI-V robustly inhibited SphK1 activation and induced accumulation of ceramides, without affecting SphK1 expression in primary OS cells. The SphK1 activator K6PC-5 or sphingosine-1-phosphate partially inhibited SKI-V-induced OS cell death. We showed that SKI-V concurrently blocked Akt-mTOR activation in primary OS cells. A constitutively-active Akt1 (ca-Akt1, S473D) construct restored Akt-mTOR activation and mitigated SKI-V-mediated cytotoxicity in primary OS cells. In vivo, daily injection of SKI-V potently suppressed OS xenograft tumor growth in nude mice. In SKI-V-administrated OS xenograft tissues, SphK1 inhibition, ceramide increase and Akt-mTOR inhibition were detected. Together, SKI-V exerts significant anti-OS activity by inhibiting SphK1 and Akt-mTOR cascades in OS cells.
Collapse
Affiliation(s)
- Xu Sun
- Department of Hand and Foot Surgery, Hospital Affiliated 5 to Nantong University, Taizhou People's Hospital, Taizhou, China
| | - Hua-Jian Shan
- Department of Orthopaedics, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Gang Yin
- Department of Orthopaedics, Wujin Hospital Affiliated to Jiangsu University, Changzhou, China
| | - Xiang-Yang Zhang
- Department of Orthopaedics, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu-Min Huang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Hai-Jun Li
- Department of Hand and Foot Surgery, Hospital Affiliated 5 to Nantong University, Taizhou People's Hospital, Taizhou, China.
| |
Collapse
|
12
|
Bu Y, Wu H, Deng R, Wang Y. Therapeutic Potential of SphK1 Inhibitors Based on Abnormal Expression of SphK1 in Inflammatory Immune Related-Diseases. Front Pharmacol 2021; 12:733387. [PMID: 34737701 PMCID: PMC8560647 DOI: 10.3389/fphar.2021.733387] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 10/04/2021] [Indexed: 01/12/2023] Open
Abstract
Sphingosine kinase 1(SphK1) a key enzyme that catalyzes the conversion of sphingosine (Sph) to sphingosine 1-phosphate (S1P), so as to maintain the dynamic balance of sphingolipid-rheostat in cells and participate in cell growth and death, proliferation and migration, vasoconstriction and remodeling, inflammation and metabolism. The normal expression of SphK1 maintains the balance of physiological and pathological states, which is reflected in the regulation of inflammatory factor secretion, immune response in traditional immune cells and non-traditional immune cells, and complex signal transduction. However, abnormal SphK1 expression and activity are found in various inflammatory and immune related-diseases, such as hypertension, atherosclerosis, Alzheimer’s disease, inflammatory bowel disease and rheumatoid arthritis. In view of the therapeutic potential of regulating SphK1 and its signal, the current research is aimed at SphK1 inhibitors, such as SphK1 selective inhibitors and dual SphK1/2 inhibitor, and other compounds with inhibitory potency. This review explores the regulatory role of over-expressed SphK1 in inflammatory and immune related-diseases, and investigate the latest progress of SphK1 inhibitors and the improvement of disease or pathological state.
Collapse
Affiliation(s)
- Yanhong Bu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, China.,College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China.,Anhui Province Key Laboratory of Research and Development of Chinese Medicine, Hefei, China
| | - Hong Wu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, China.,College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China.,Anhui Province Key Laboratory of Research and Development of Chinese Medicine, Hefei, China
| | - Ran Deng
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, China.,College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China.,Anhui Province Key Laboratory of Research and Development of Chinese Medicine, Hefei, China
| | - Yan Wang
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, China.,College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China.,Anhui Province Key Laboratory of Research and Development of Chinese Medicine, Hefei, China
| |
Collapse
|
13
|
Jiang Y, Gu C, Xu H, Shi F, Zhang X, Wang F. iKeap1 activates Nrf2 signaling to protect myocardial cells from oxygen glucose deprivation/re-oxygenation-induced oxidative injury. Biochem Biophys Res Commun 2021; 574:110-117. [PMID: 34461498 DOI: 10.1016/j.bbrc.2021.08.024] [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: 07/05/2021] [Revised: 08/05/2021] [Accepted: 08/07/2021] [Indexed: 11/24/2022]
Abstract
Nuclear factor E2-related factor 2 (Nrf2) activation could efficiently protect myocardial cells from oxygen glucose deprivation/re-oxygenation (OGDR). An ultra-large structure-based virtual screening has discovered iKeap1 as a novel, direct and potent Keap1 inhibitor. Here we found that iKeap1 efficiently activated Nrf2 signaling in H9c2 myocardial cells and primary murine myocardiocytes. iKeap1 induced Keap1-Nrf2 disassociation, cytosol Nrf2 protein stabilization and nuclear translocation. The antioxidant response element (ARE) activity and expression of Nrf2 cascade genes (HO1, NQO1 and GCLC) were increased in iKeap1-treated myocardial cells. In H9c2 cells and murine myocardiocytes, iKeap1 potently inhibited OGDR-induced oxidative injury by inhibiting reactive oxygen species (ROS) production, mitochondrial depolarization, lipid peroxidation and DNA damage. In addition, OGDR-induced myocardial cell death and apoptosis were largely ameliorated after pretreatment with the novel Keap1 inhibitor. Significantly, in H9c2 cells iKeap1-induced myocardial cytoprotection against OGDR was abolished with Nrf2 silencing or knockout (using CRISPR/Cas9 method). Moreover, CRISPR/Cas9-induced Keap1 knockout led to constitutive activation of Nrf2 cascade and inhibited OGDR-induced oxidative injury. Importantly, iKeap1 was unable to further protect Keap1-knockout H9c2 cells from OGDR. Together, iKeap1 activated Nrf2 signaling to protect myocardial cells from OGDR-induced oxidative injury and cell death.
Collapse
Affiliation(s)
- Yicheng Jiang
- Department of Cardiology, Huai'an First People's Hospital, Nanjing Medical University, China
| | - Cheng Gu
- Department of Critical Care Medicine, Changshu No.2 People's Hospital, Changshu, China
| | - Hai Xu
- Department of Cardiology, Huai'an First People's Hospital, Nanjing Medical University, China
| | - Feiya Shi
- Department of Cardiology, Huai'an First People's Hospital, Nanjing Medical University, China
| | - Xiwen Zhang
- Department of Cardiology, Huai'an First People's Hospital, Nanjing Medical University, China.
| | - Fang Wang
- Department of Cardiology, Shanghai General Hospital of Nanjing Medical University, Shanghai, China.
| |
Collapse
|
14
|
Imre G, Krähling V, Eichler M, Trautmann S, Ferreirós N, Aman MJ, Kashanchi F, Rajalingam K, Pöhlmann S, Becker S, Meyer Zu Heringdorf D, Pfeilschifter J. The sphingosine kinase 1 activator, K6PC-5, attenuates Ebola virus infection. iScience 2021; 24:102266. [PMID: 33817572 PMCID: PMC8005759 DOI: 10.1016/j.isci.2021.102266] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 02/08/2021] [Accepted: 03/01/2021] [Indexed: 12/18/2022] Open
Abstract
Ebola virus (EBOV) is responsible for outbreaks with case fatality rates of up to 90% and for an epidemic in West Africa with more than ten thousand deaths. EBOV glycoprotein (EBOV-GP) is the only viral surface protein and is responsible for viral entry into cells. Here, by employing pseudotyped EBOV-GP viral particles, we uncover a critical role for sphingolipids in inhibiting viral entry. Sphingosine kinase 1 (SphK1) catalyzes the phosphorylation of sphingosine to sphingosine 1-phosphate (S1P). The administration of the SphK1 activator, K6PC-5, or S1P, or the overexpression of SphK1 consistently exhibited striking inhibitory effects in EBOV-GP-driven entry in diverse cell lines. Finally, K6PC-5 markedly reduced the EBOV titer in infected cells and the de novo production of viral proteins. These data present K6PC-5 as an efficient tool to inhibit EBOV infection in endothelial cells and suggest further studies to evaluate its systemic effects. K6PC-5, a sphingosine kinase 1 activator, inhibits Ebola virus infection Sphingosine 1-phosphate, the product of SphK1, attenuates the viral entry Inhibiton/activation of S1P receptors has no influence on Ebola virus entry These data support the endogen effect of S1P in Ebola virus infection
Collapse
Affiliation(s)
- Gergely Imre
- Institute of General Pharmacology and Toxicology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main 60590, Germany
| | - Verena Krähling
- Institute of Virology, Philipps University Marburg, Marburg, Germany.,German Center for Infection Research (DZIF), partner site Gießen-Marburg-Langen, Marburg, Germany
| | - Madeleine Eichler
- Institute of General Pharmacology and Toxicology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main 60590, Germany
| | - Sandra Trautmann
- Institute of Clinical Pharmacology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main 60590, Germany
| | - Nerea Ferreirós
- Institute of Clinical Pharmacology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main 60590, Germany
| | - M Javad Aman
- Integrated BioTherapeutics, Inc., Gaithersburg, MD 20850, USA
| | - Fatah Kashanchi
- Laboratory of Molecular Virology, George Mason University Manassas, VA 20110, USA
| | - Krishnaraj Rajalingam
- Cell Biology Unit, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Stefan Pöhlmann
- Infection Biology Unit, German Primate Center - Leibniz Institute for Primate Research, 37077 Göttingen, Germany.,Faculty of Biology and Psychology, University Göttingen, 37077 Göttingen, Germany
| | - Stephan Becker
- Institute of Virology, Philipps University Marburg, Marburg, Germany.,German Center for Infection Research (DZIF), partner site Gießen-Marburg-Langen, Marburg, Germany
| | - Dagmar Meyer Zu Heringdorf
- Institute of General Pharmacology and Toxicology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main 60590, Germany
| | - Josef Pfeilschifter
- Institute of General Pharmacology and Toxicology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main 60590, Germany
| |
Collapse
|
15
|
Xu HB, Zheng YF, Wu D, Li Y, Zhou LN, Chen YG. microRNA-1203 targets and silences cyclophilin D to protect human endometrial cells from oxygen and glucose deprivation-re-oxygenation. Aging (Albany NY) 2020; 12:3010-3024. [PMID: 32041924 PMCID: PMC7041737 DOI: 10.18632/aging.102795] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 01/12/2020] [Indexed: 02/06/2023]
Abstract
Oxygen and glucose deprivation (OGD)-re-oxygenation (OGDR) stimulation to the human endometrial cells mimics ischemia-reperfusion injury. Cyclophilin D (CypD)-dependent programmed necrosis pathway mediates OGDR-induced cytotoxicity to human endometrial cells. We here identified a novel CypD-targeting miRNA, microRNA-1203 (miR-1203). In T-HESC and primary human endometrial cells, ectopic overexpression of miR-1203, using a lentiviral construct, potently downregulated the CypD 3’-untranslated region (3’-UTR) activity and its expression. Both were however upregulated in endometrial cells with forced miR-1203 inhibition by its anti-sense sequence. Functional studies demonstrated that ectopic miR-1203 overexpression in endometrial cells alleviated OGDR-induced programmed necrosis, inhibiting mitochondrial CypD-p53-adenine nucleotide translocator 1 association, mitochondrial depolarization, reactive oxygen species production, and medium lactate dehydrogenase release. Contrarily OGDR-induced programmed necrosis and cytotoxicity were intensified with forced miR-1203 inhibition in endometrial cells. Significantly, ectopic miR-1203 overexpression or inhibition failed to change OGDR-induced cytotoxicity in CypD-knockout T-HESC cells. Furthermore, ectopic miR-1203 overexpression was unable to protect T-HESC endometrial cells from OGDR when CypD was restored by an UTR-depleted CypD construct. Collectively, these results show that miR-1203 targets and silences CypD to protect human endometrial cells from OGDR
Collapse
Affiliation(s)
- Hong-Bin Xu
- Obstetrics and Gynecology Department, The First Affiliated Hospital of Soochow University, Suzhou, China.,Obstetrics and Gynecology Department, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Yu-Fan Zheng
- Institute of Neuroscience, Soochow University, Suzhou, China
| | - Di Wu
- Institute of Neuroscience, Soochow University, Suzhou, China
| | - Ya Li
- The Central Lab, North District, Suzhou Municipal Hospital Affiliated to Nanjing Medical University, Suzhou, China
| | - Li-Na Zhou
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Suzhou, China
| | - You-Guo Chen
- Obstetrics and Gynecology Department, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
16
|
GSK621 attenuates oxygen glucose deprivation/re-oxygenation-induced myocardial cell injury via AMPK-dependent signaling. Biochem Biophys Res Commun 2019; 514:826-834. [DOI: 10.1016/j.bbrc.2019.04.196] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 04/29/2019] [Indexed: 12/25/2022]
|
17
|
Shi X, Liu HY, Li SP, Xu HB. Keratinocyte growth factor protects endometrial cells from oxygen glucose deprivation/re-oxygenation via activating Nrf2 signaling. Biochem Biophys Res Commun 2018; 501:178-185. [DOI: 10.1016/j.bbrc.2018.04.208] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 04/26/2018] [Indexed: 12/20/2022]
|
18
|
Lin B, Xu J, Feng DG, Wang F, Wang JX, Zhao H. DUSP14 knockout accelerates cardiac ischemia reperfusion (IR) injury through activating NF-κB and MAPKs signaling pathways modulated by ROS generation. Biochem Biophys Res Commun 2018; 501:24-32. [DOI: 10.1016/j.bbrc.2018.04.101] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 04/13/2018] [Indexed: 11/25/2022]
|
19
|
Law BA, Hancock WD, Cowart LA. Getting to the heart of the sphingolipid riddle. CURRENT OPINION IN PHYSIOLOGY 2018; 1:111-122. [PMID: 33195889 PMCID: PMC7665081 DOI: 10.1016/j.cophys.2017.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Obesity, Type 2 Diabetes, and Metabolic Syndrome induce dyslipidemia resulting in inundation of peripheral organs with fatty acids. These not only serve as substrates for energy production, but also contribute to aberrant production of bioactive lipids. Moreover, lipid metabolism is affected in many cardiac disorders including heart failure, ischemia reperfusion injury, and others. While lipids serve crucial homeostatic roles, perturbing biosynthesis of lipid mediators leads to aberrant cell signaling, which contributes to maladaptive cardiovascular programs. Bioactive sphingolipids, in particular, have been implicated in pathophysiology in the heart and vasculature by a variety of studies in cells, animal models, and humans. Because of the burgeoning interest in sphingolipid-driven biology in the cardiovascular system, it is necessary to discuss the experimental considerations for studying sphingolipid metabolism and signaling, emphasizing the caveats to some widely available experimental tools and approaches. Additionally, there is a growing appreciation for the diversity of ceramide structures generated via specific enzymes and bearing disparate cellular functions. While targeting these individual species and enzymes constitutes a major advance, studies show that sphingolipid synthesis readily adapts to compensate for experimental targeting of any individual pathway, thereby convoluting data interpretation. Furthermore, though some molecular mechanisms of sphingolipid action are known, signaling pathways impacted by sphingolipids remain incompletely understood. In this review, we discuss these issues and highlight recent studies as well as future directions that may extend our understanding of the metabolism and signaling actions of these enigmatic lipids in the cardiovascular context.
Collapse
Affiliation(s)
- Britany A Law
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC
- Present Address: Department of Medicine-Cardiology, Duke University, Durham NC
| | - William D Hancock
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC
| | - L Ashley Cowart
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC
- Ralph H. Johnson Veteran's Affairs Medical Center, Charleston, SC
| |
Collapse
|
20
|
Zheng J, Cui E, Yang H, Li M, Zhou J, Yan M, Sun J, Tang DR. Targeting cyclophilin-D by compound 19 protects neuronal cells from oxygen glucose deprivation/re-oxygenation. Oncotarget 2017; 8:90238-90249. [PMID: 29163824 PMCID: PMC5685745 DOI: 10.18632/oncotarget.21655] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 08/28/2017] [Indexed: 01/03/2023] Open
Abstract
Oxygen and glucose deprivation (OGD) with re-oxygenation (OGDR) is applied to neuronal cells to mimic ischemia-reperfusion injuries. Activation of cyclophilin D (Cyp-D)-dependent programmed necrosis pathway mediates OGDR-induced neuronal cell damages. Here, we tested the potential effect of Compound 19 (C19), a novel Cyp-D inhibitor, in this process. In both established neuronal cell lines (Neuro-2a and NB41A3 cells) and the primary murine CA1 hippocampal neurons, pretreatment with C19 largely attenuated OGDR-induced cell viability reduction and cell death. Significantly, C19 was ineffective in Cyp-D-silenced Neuro-2a cells. OGDR induced mitochondria-dependent programmed necrosis in neuronal cells. OGDR induced p53 translocation to mitochondria and association with Cyp-D, causing mitochondrial depolarization, cytochrome C release and reactive oxygen species production. Such effects were largely attenuated with pre-treatment of C19. Importantly, C19 was significantly more efficient than other known Cyp-D inhibitors in protecting neuronal cells from OGDR. These results suggest that targeting Cyp-D by C19 protects neuronal cells from OGDR.
Collapse
Affiliation(s)
- Jinyu Zheng
- Department of Neurosurgery, The Affiliated Huai'an Hospital of Xuzhou Medical College, Huai'an, China
| | - Enhui Cui
- Department of Anesthesiology, Huai'an Maternity and Child Healthcare Hospital, Yangzhou University Medical School, Huai'an, China
| | - Haikou Yang
- Department of Anesthesiology, Huai'an Maternity and Child Healthcare Hospital, Yangzhou University Medical School, Huai'an, China
| | - Mao Li
- Department of Anesthesiology, Huai'an Maternity and Child Healthcare Hospital, Yangzhou University Medical School, Huai'an, China
| | - Jing Zhou
- Department of Anesthesiology, Huai'an Maternity and Child Healthcare Hospital, Yangzhou University Medical School, Huai'an, China
| | - Ming Yan
- Department of Anesthesiology, Huai'an Maternity and Child Healthcare Hospital, Yangzhou University Medical School, Huai'an, China
| | - Jian Sun
- Department of Anesthesiology, Huai'an Maternity and Child Healthcare Hospital, Yangzhou University Medical School, Huai'an, China
| | - De-Rong Tang
- Department of Thoracic Surgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, China
| |
Collapse
|
21
|
Zheng K, Zhang Q, Lin G, Li Y, Sheng Z, Wang J, Chen L, Lu HH. Activation of Akt by SC79 protects myocardiocytes from oxygen and glucose deprivation (OGD)/re-oxygenation. Oncotarget 2017; 8:14978-14987. [PMID: 28122357 PMCID: PMC5362459 DOI: 10.18632/oncotarget.14785] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 01/11/2017] [Indexed: 12/21/2022] Open
Abstract
SC79 is a novel Akt activator. The current study tested its potential effect against oxygen and glucose deprivation (OGD)/re-oxygenation-induced myocardial cell death. We showed that SC79 activated Akt and protected H9c2 myocardial cells and primary murine myocardiocytes from OGD/re-oxygenation. Reversely, Akt inhibitor MK-2206 or Akt1 shRNA knockdown almost completely abolished SC79-mediated myocardial cytoprotection. SC79 treatment in H9c2 cells inhibited OGD/re-oxygenation-induced programmed necrosis pathway, evidenced by mitochondrial depolarization and cyclophilin D-p53-ANT-1 (adenine nucleotide translocator 1) association. Further, SC79 activated Akt downstream NF-E2-related factor 2 (NRF2) signaling to suppress OGD/re-oxygenation-induced reactive oxygen species (ROS) production. Reversely, NRF2 shRNA knockdown in H9c2 cells largely attenuated SC79-induced ROS scavenging ability and cytoprotection against OGD/re-oxygenation. Together, we conclude that activation of Akt by SC79 protects myocardial cells from OGD/re-oxygenation.
Collapse
Affiliation(s)
- Koulong Zheng
- Department of Cardiology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Qing Zhang
- Department of Cardiology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Gang Lin
- Department of Cardiology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Yefei Li
- Department of Cardiology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Zhenqiang Sheng
- Department of Cardiology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Jue Wang
- Department of Cardiology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Liang Chen
- Department of Cardiology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Hui-He Lu
- Department of Cardiology, The Second Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
22
|
Defective Sphingosine-1-phosphate metabolism is a druggable target in Huntington's disease. Sci Rep 2017; 7:5280. [PMID: 28706199 PMCID: PMC5509685 DOI: 10.1038/s41598-017-05709-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 06/01/2017] [Indexed: 12/22/2022] Open
Abstract
Huntington’s disease is characterized by a complex and heterogeneous pathogenic profile. Studies have shown that disturbance in lipid homeostasis may represent a critical determinant in the progression of several neurodegenerative disorders. The recognition of perturbed lipid metabolism is only recently becoming evident in HD. In order to provide more insight into the nature of such a perturbation and into the effect its modulation may have in HD pathology, we investigated the metabolism of Sphingosine-1-phosphate (S1P), one of the most important bioactive lipids, in both animal models and patient samples. Here, we demonstrated that S1P metabolism is significantly disrupted in HD even at early stage of the disease and importantly, we revealed that such a dysfunction represents a common denominator among multiple disease models ranging from cells to humans through mouse models. Interestingly, the in vitro anti-apoptotic and the pro-survival actions seen after modulation of S1P-metabolizing enzymes allows this axis to emerge as a new druggable target and unfolds its promising therapeutic potential for the development of more effective and targeted interventions against this incurable condition.
Collapse
|
23
|
The novel cyclophilin D inhibitor compound 19 protects retinal pigment epithelium cells and retinal ganglion cells from UV radiation. Biochem Biophys Res Commun 2017; 487:807-812. [DOI: 10.1016/j.bbrc.2017.04.128] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 04/23/2017] [Indexed: 12/23/2022]
|
24
|
Zhang S, Deng Z, Yao C, Huang P, Zhang Y, Cao S, Li X. AT7867 Inhibits Human Colorectal Cancer Cells via AKT-Dependent and AKT-Independent Mechanisms. PLoS One 2017; 12:e0169585. [PMID: 28081222 PMCID: PMC5231330 DOI: 10.1371/journal.pone.0169585] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 12/19/2016] [Indexed: 11/18/2022] Open
Abstract
AKT is often hyper-activated in human colorectal cancers (CRC). This current study evaluated the potential anti-CRC activity by AT7867, a novel AKT and p70S6K1 (S6K1) dual inhibitor. We showed that AT7867 inhibited survival and proliferation of established (HT-29, HCT116 and DLD-1 lines) and primary human CRC cells. Meanwhile, it provoked caspase-dependent apoptosis in the CRC cells. Molecularly, AT7867 blocked AKT-S6K1 activation in CRC cells. Restoring AKT-S6K1 activation, via expression of a constitutively-active AKT1 ("ca-AKT1"), only partially attenuated AT7867-induced HT-29 cell death. Further studies demonstrated that AT7867 inhibited sphingosine kinase 1 (SphK1) activity to promote pro-apoptotic ceramide production in HT-29 cells. Such effects by AT7867 were independent of AKT inhibition. AT7867-indued ceramide production and subsequent HT-29 cell apoptosis were attenuated by co-treatment of sphingosine-1-phosphate (S1P), but were potentiated with the glucosylceramide synthase (GCS) inhibitor PDMP. In vivo, intraperitoneal injection of AT7867 inhibited HT-29 xenograft tumor growth in nude mice. AKT activation was also inhibited in AT7867-treated HT-29 tumors. Together, the preclinical results suggest that AT7867 inhibits CRC cells via AKT-dependent and -independent mechanisms.
Collapse
Affiliation(s)
- Shihu Zhang
- Department of General Surgery, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhengming Deng
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Chen Yao
- Orthopedic Department, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Ping Huang
- Department of General Surgery, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yi Zhang
- Department of General Surgery, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shibing Cao
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiangcheng Li
- Department of General Surgery, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
25
|
Su G, Zhang R, Yang X, Bai R, Yin X, Gao X, Li L, Tu P, Chai X. Lignans from the stem bark of Syringa pinnatifolia. Fitoterapia 2016; 114:63-68. [PMID: 27568323 DOI: 10.1016/j.fitote.2016.08.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 08/18/2016] [Accepted: 08/20/2016] [Indexed: 01/04/2023]
Abstract
Four new lignans, alashinols A-D (1-4), a new hydrolysis product, alashinols E (5), and seven known analogues were isolated from the stem bark of Syringa pinnatifolia Hemsl. These new lignans were characterized using 1D and 2D NMR and MS data, and their absolute configurations were determined by experimental and calculated electronic circular dichroism and X-ray diffraction analyses. Alashinol B (2) exhibited two conformers that adopted an unusual unit cell packing. Anti-inflammatory evaluation revealed that compounds 1, 4, 6, and 8 showed moderate inhibition against NO production in lipopolysaccharide-induced macrophages RAW 264.7 cells with IC50 values range from 43.3-60.9μM, and 1 decreased the TNF-α and IL-6 level in a concentration-dependent manner at 40-160μM and exhibited considerable neuroprotective effect against the glutamate-induced injury in PC12 cell line. Analogues 3 and 9 showed protective effects against oxygen glucose deprivation/reoxygenation in H9c2 cells.
Collapse
Affiliation(s)
- Guozhu Su
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, People's Republic of China; School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100102, People's Republic of China
| | - Ruifei Zhang
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, People's Republic of China; School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100102, People's Republic of China
| | - Xinyao Yang
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, People's Republic of China; School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100102, People's Republic of China
| | - Ruifeng Bai
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, People's Republic of China; School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100102, People's Republic of China
| | - Xu Yin
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, People's Republic of China; School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100102, People's Republic of China
| | - Xiaoli Gao
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, People's Republic of China
| | - Li Li
- Department of Medicinal Chemistry, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Pengfei Tu
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, People's Republic of China
| | - Xingyun Chai
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, People's Republic of China.
| |
Collapse
|
26
|
Xu L, Zhang Y, Gao M, Wang G, Fu Y. Concurrent targeting Akt and sphingosine kinase 1 by A-674563 in acute myeloid leukemia cells. Biochem Biophys Res Commun 2016; 472:662-8. [PMID: 26920060 DOI: 10.1016/j.bbrc.2016.02.094] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 02/22/2016] [Indexed: 12/13/2022]
Abstract
Akt signaling plays a pivotal role in acute myeloid leukemia (AML) development and progression. In the present study, we evaluated the potential anti-AML activity by a novel Akt kinase inhibitor A-674563. Our results showed that A-674563 dose-dependently inhibited survival and proliferation of U937 AML cells and six lines of human AML progenitor cells, yet sparing human peripheral blood mononuclear leukocytes (PBMCs). A-674563 activated caspase-3/9 and apoptosis in the AML cells. Reversely, the pan-caspase inhibitor z-VAD-CHO dramatically alleviated A-674563-induced AML cell apoptosis and cytotoxicity. For the molecular study, we showed that A-674563 blocked Akt activation in U937 cells and human AML progenitor cells. Further, A-674563 decreased sphingosine kinase 1 (SphK1) activity in above AML cells to deplete pro-survival sphingosine-1-phosphate (S1P) and boost pro-apoptotic ceramide production. Such an effect on SphK1 signaling by A-674563 appeared independent of Akt blockage. Significantly, K6PC-5, a novel SphK1 activator, or supplement with S1P attenuated A-674563-induced ceramide production, and subsequent U937 cell death and apoptosis. Importantly, intraperitoneal injection of A-674563 at well-tolerated doses suppressed U937 leukemic xenograft tumor growth in nude mice, whiling significantly improving the animal survival. The results of the current study demonstrate that A-674563 exerts potent anti-leukemic activity in vitro and in vivo, possibly via concurrent targeting Akt and SphK1 signalings.
Collapse
Affiliation(s)
- Lin Xu
- Xiangya Hospital, Central South University, Changsha, China; Shaoyang Central Hospital, Hunan Province, China
| | - Yanan Zhang
- The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Meng Gao
- The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Guangping Wang
- Xiangya Hospital, Central South University, Changsha, China.
| | - Yunfeng Fu
- The Third Xiangya Hospital, Central South University, Changsha, 410013, China.
| |
Collapse
|
27
|
Zheng K, Lu H, Sheng Z, Li Y, Xu B. Low-concentration of perifosine surprisingly protects cardiomyocytes from oxygen glucose deprivation. Biochem Biophys Res Commun 2015; 469:753-60. [PMID: 26686418 DOI: 10.1016/j.bbrc.2015.12.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 12/02/2015] [Indexed: 12/25/2022]
Abstract
Here we found that low-concentration of perifosine, an Akt inhibitor, surprisingly protected cardiomyocytes from oxygen glucose deprivation (OGD)/re-oxygenation. In H9c2 cardiomyocytes, non-cytotoxic perifosine (0.1-0.5 μM) suppressed OGD/re-oxygenation-induced reactive oxygen species (ROS) production, p53 mitochondrial translocation and cyclophilin D complexation, as well as mitochondrial membrane potential (MMP) reduction. Molecularly, perifosine activated AMP-activated kinase (AMPK) signaling to increase intracellular NADPH (nicotinamide adenine dinucleotide phosphate) content in H9c2 cells. On the other hand, AMPK inhibition by AMPKα1 shRNA-knockdown in H9c2 cells significantly reduced perifosine-induced NADPH production, and alleviated perifosine-mediated anti-oxidant and cytoprotective activities against OGD/re-oxygenation. In primary murine cardiomyocytes, perifosine similarly activated AMPK signaling, and offered significant protection against OGD/re-oxygenation, which was largely attenuated with siRNA knockdown of AMPKα1. We demonstrate an unexpected function of perifosine (low-concentration) in protecting cardiomyocytes from OGD/re-oxygenation.
Collapse
Affiliation(s)
- Koulong Zheng
- Department of Cardiology, Drum Tower Clinical Medical Hospital, Nanjing Medical University, Nanjing, China; Department of Cardiology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Huihe Lu
- Department of Cardiology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Zhenqiang Sheng
- Department of Cardiology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Yefei Li
- Department of Cardiology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Biao Xu
- Department of Cardiology, Drum Tower Clinical Medical Hospital, Nanjing Medical University, Nanjing, China.
| |
Collapse
|