1
|
Brunmaier LAE, Ozdemir T, Walker TW. Angiogenesis: Biological Mechanisms and In Vitro Models. Ann Biomed Eng 2025:10.1007/s10439-025-03721-2. [PMID: 40210793 DOI: 10.1007/s10439-025-03721-2] [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] [Received: 01/10/2025] [Accepted: 03/25/2025] [Indexed: 04/12/2025]
Abstract
The translation of biomedical devices and drug research is an expensive and long process with a low probability of receiving FDA approval. Developing physiologically relevant in vitro models with human cells offers a solution to not only improving the odds of FDA approval but also to expand our ability to study complex in vivo systems in a simpler fashion. Animal models remain the standard for pre-clinical testing; however, the data from animal models is an unreliable extrapolation when anticipating a human response in clinical trials, thus contributing to the low rates of translation. In this review, we focus on in vitro vascular or angiogenic models because of the incremental role that the vascular system plays in the translation of biomedical research. The first section of this review discusses the most common angiogenic cytokines that are used in vitro to initiate angiogenesis, followed by angiogenic inhibitors where both initiators and inhibitors work to maintain vascular homeostasis. Next, we evaluate previously published in vitro models, where we evaluate capturing the physical environment for biomimetic in vitro modeling. These topics provide a foundation of parameters that must be considered to improve and achieve vascular biomimicry. Finally, we summarize these topics to suggest a path forward with the goal of engineering human in vitro models that emulate the in vivo environment and provide a platform for biomedical device and drug screening that produces data to support clinical translation.
Collapse
Affiliation(s)
- Laura A E Brunmaier
- Nanoscience and Biomedical Engineering Department, South Dakota School of Mines & Technology, 501 E St. Joseph St., Rapid City, SD, 57701, USA
| | - Tugba Ozdemir
- Nanoscience and Biomedical Engineering Department, South Dakota School of Mines & Technology, 501 E St. Joseph St., Rapid City, SD, 57701, USA
| | - Travis W Walker
- Karen M. Swindler Department of Chemical and Biological Engineering, South Dakota School of Mines & Technology, 501 E St. Joseph St., Rapid City, SD, 57701, USA.
| |
Collapse
|
2
|
Zhang J, Ge Q, Du T, Kuang Y, Fan Z, Jia X, Gu W, Chen Z, Wei Z, Shen B. SPHK1/S1PR1/PPAR-α axis restores TJs between uroepithelium providing new ideas for IC/BPS treatment. Life Sci Alliance 2025; 8:e202402957. [PMID: 39578076 PMCID: PMC11584326 DOI: 10.26508/lsa.202402957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 11/13/2024] [Accepted: 11/15/2024] [Indexed: 11/24/2024] Open
Abstract
Interstitial cystitis/bladder pain syndrome (IC/BPS) represents a chronic, aseptic inflammatory bladder condition with an unclear etiology and few therapeutic options. A composite barrier structure composed of the uroepithelium and glycosaminoglycan layer forms on the bladder's inner surface to block urine and other harmful substances. Dysfunction of this barrier may initiate the pathogenesis of IC/BPS. Sphingosine-1-phosphate (S1P) plays a crucial role in forming tight junctions. Perfusion of S1P into the bladder restored uroepithelial tight junctions in mice with cyclophosphamide-induced acute cystitis and ameliorated symptoms of the lower urinary tract. Mice lacking sphingosine kinase 1 (SHPK1) exhibited more severe bladder injuries and dysfunction. Concurrent in vitro experiments elucidated S1P's protective effects and its role as a primary messenger through SPHK1 and S1P receptor 1 (S1PR1) knockdown. This study identifies a novel mechanism whereby S1P binding to S1PR1 activates the PPAR-α pathway, thereby enhancing cholesterol transport and restoring tight junctions between uroepithelial cells. These findings elucidate the regulatory role of S1P in the bladder epithelial barrier and highlight a promising therapeutic target for IC/BPS.
Collapse
Affiliation(s)
- Junjie Zhang
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Urology, The Second Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Qingyu Ge
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Urology, The Second Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Tianpeng Du
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Urology, The Second Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Yuhao Kuang
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Urology, The Second Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Zongyao Fan
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Urology, The Second Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Xinyi Jia
- Respiratory Department, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Wenjin Gu
- Nanjing Medical University, Nanjing, China
| | - Zhengsen Chen
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Urology, The Second Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Zhongqing Wei
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Urology, The Second Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Baixin Shen
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Urology, The Second Clinical Medical College of Nanjing Medical University, Nanjing, China
| |
Collapse
|
3
|
Wang L, Kong Q, Leng X, Leung H, Li Y. The sphingosine-1-phosphate signaling pathway (sphingosine-1-phosphate and its receptor, sphingosine kinase) and epilepsy. Epilepsia Open 2025; 10:55-73. [PMID: 39727628 PMCID: PMC11803289 DOI: 10.1002/epi4.13112] [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: 04/15/2024] [Revised: 11/08/2024] [Accepted: 11/20/2024] [Indexed: 12/28/2024] Open
Abstract
Epilepsy is one of the common chronic neurological diseases, affecting more than 70 million people worldwide. The brains of people with epilepsy exhibit a pathological and persistent propensity for recurrent seizures. Epilepsy often coexists with cardiovascular disease, cognitive dysfunction, depression, etc., which seriously affects the patient's quality of life. Although our understanding of epilepsy has advanced, the pathophysiological mechanisms leading to epileptogenesis, drug resistance, and associated comorbidities remain largely unknown. The use of newer antiepileptic drugs has increased, but this has not improved overall outcomes. We need to deeply study the pathogenesis of epilepsy and find drugs that can not only prevent the epileptogenesis and interfere with the process of epileptogenesis but also treat epilepsy comorbidities. Sphingosine-1-phosphate (S1P) is an important lipid molecule. It not only forms the basis of cell membranes but is also an important bioactive mediator. It can not only act as a second messenger in cells to activate downstream signaling pathways but can also exert biological effects by being secreted outside cells and binding to S1P receptors on the cell membrane. Fingolimod (FTY720) is the first S1P receptor modulator developed and approved for the treatment of multiple sclerosis. More and more studies have proven that the S1P signaling pathway is closely related to epilepsy, drug-resistant epilepsy, epilepsy comorbidities, or other epilepsy-causing diseases. However, there is much controversy over the role of certain natural molecules in the pathway and receptor modulators (such as FTY720) in epilepsy. Here, we summarize and analyze the role of the S1P signaling pathway in epilepsy, provide a basis for finding potential therapeutic targets and/or epileptogenic biomarkers, analyze the reasons for these controversies, and put forward our opinions. PLAIN LANGUAGE SUMMARY: This article combines the latest research literature at home and abroad to review the sphingosine 1-phosphate signaling pathway and epileptogenesis, drug-resistant epilepsy, epilepsy comorbidities, other diseases that can cause epilepsy, as well as the sphingosine-1-phosphate signaling pathway regulators and epilepsy, with the expectation of providing a certain theoretical basis for finding potential epilepsy treatment targets and/or epileptogenic biomarkers in the sphingosine-1-phosphate signaling pathway.
Collapse
Affiliation(s)
- Lin Wang
- Department of NeurologyAffiliated Hospital of Jining Medical UniversityJining CityChina
- Epilepsy CenterAffiliated Hospital of Jining Medical UniversityJining CityChina
- The Chinese University of Hong Kong, Department of Medicine and TherapeuticsThe Chinese University of Hong Kong, Central AveHong KongHong Kong
| | - Qingxia Kong
- Department of NeurologyAffiliated Hospital of Jining Medical UniversityJining CityChina
- Epilepsy CenterAffiliated Hospital of Jining Medical UniversityJining CityChina
| | - Xinyi Leng
- The Chinese University of Hong Kong, Department of Medicine and TherapeuticsThe Chinese University of Hong Kong, Central AveHong KongHong Kong
| | - Howan Leung
- Division of Neurology, Department of Medicine and Therapeutics, Prince of Wales Hospital7/F Clinical Science Building, Prince of Wales HospitalHong KongHong Kong
| | - Yang Li
- Department of OncologyAffiliated Hospital of Jining Medical UniversityJining CityChina
| |
Collapse
|
4
|
Li HZ, Pike ACW, Chang YN, Prakaash D, Gelova Z, Stanka J, Moreau C, Scott HC, Wunder F, Wolf G, Scacioc A, McKinley G, Batoulis H, Mukhopadhyay S, Garofoli A, Pinto-Fernández A, Kessler BM, Burgess-Brown NA, Štefanić S, Wiedmer T, Dürr KL, Puetter V, Ehrmann A, Khalid S, Ingles-Prieto A, Superti-Furga G, Sauer DB. Transport and inhibition of the sphingosine-1-phosphate exporter SPNS2. Nat Commun 2025; 16:721. [PMID: 39820269 PMCID: PMC11739509 DOI: 10.1038/s41467-025-55942-7] [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: 04/02/2024] [Accepted: 01/03/2025] [Indexed: 01/19/2025] Open
Abstract
Sphingosine-1-phosphate (S1P) is a signaling lysolipid critical to heart development, immunity, and hearing. Accordingly, mutations in the S1P transporter SPNS2 are associated with reduced white cell count and hearing defects. SPNS2 also exports the S1P-mimicking FTY720-P (Fingolimod) and thereby is central to the pharmacokinetics of this drug when treating multiple sclerosis. Here, we use a combination of cryo-electron microscopy, immunofluorescence, in vitro binding and in vivo S1P export assays, and molecular dynamics simulations to probe SPNS2's substrate binding and transport. These results reveal the transporter's binding mode to its native substrate S1P, the therapeutic FTY720-P, and the reported SPNS2-targeting inhibitor 33p. Further capturing an inward-facing apo state, our structures illuminate the protein's mechanism for exchange between inward-facing and outward-facing conformations. Finally, using these structural, localization, and S1P transport results, we identify how pathogenic mutations ablate the protein's export activity and thereby lead to hearing loss.
Collapse
Affiliation(s)
- Huanyu Z Li
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Ashley C W Pike
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | | | - Zuzana Gelova
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | | | - Christophe Moreau
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Hannah C Scott
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Chinese Academy for Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | - Gernot Wolf
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Andreea Scacioc
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Gavin McKinley
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | - Shubhashish Mukhopadhyay
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Andrea Garofoli
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Adán Pinto-Fernández
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Chinese Academy for Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Benedikt M Kessler
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Chinese Academy for Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Nicola A Burgess-Brown
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Saša Štefanić
- Nanobody Service Facility, University of Zurich, AgroVet-Strickhof, Lindau, Switzerland
| | - Tabea Wiedmer
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Katharina L Dürr
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| | | | | | - Syma Khalid
- Department of Biochemistry, University of Oxford, Oxford, UK.
| | - Alvaro Ingles-Prieto
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.
| | - Giulio Superti-Furga
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria.
| | - David B Sauer
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
5
|
Liu X, Yue J, Zhou C, Duan Y, Chen X, Liu J, Zhuang S, Luo Y, Wu J, Zhang Y, Zhang L. Cardiomyocyte S1PR1 promotes cardiac regeneration via AKT/mTORC1 signaling pathway. Theranostics 2025; 15:1524-1551. [PMID: 39816679 PMCID: PMC11729560 DOI: 10.7150/thno.103797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 12/09/2024] [Indexed: 01/18/2025] Open
Abstract
Aims: Lower vertebrates and some neonatal mammals are known to possess the ability to regenerate cardiomyocyte and fully recover after heart injuries within a limited period. Understanding the molecular mechanisms of heart regeneration and exploring new ways to enhance cardiac regeneration hold significant promise for therapeutic intervention of heart failure. Sphingosine 1-phospahte receptor 1 (S1PR1) is highly expressed in cardiomyocytes and plays a crucial role in heart development and pathological cardiac remodeling. However, the effect of cardiomyocyte-expressing S1PR1 on heart regeneration has not yet been elucidated. This study aims to investigate the role of cardiomyocyte S1PR1 in cardiac regeneration following heart injuries. Methods and Results: We generated cardiomyocyte (CM)-specific S1pr1 knock-out mice and demonstrated that CM-specific S1pr1 loss-of-function severely reduced cardiomyocyte proliferation and inhibited heart regeneration following apex resection in neonatal mice. Conversely, AAV9-mediated CM-specific S1pr1 gain-of-function significantly enhanced cardiac regeneration. We identified that S1PR1 activated the AKT/mTORC1/CYCLIN D1 and BCL2 signaling pathways to promote cardiomyocyte proliferation and inhibit apoptosis. Moreover, CM-targeted gene delivery system via AAV9 to overexpress S1PR1 significantly increased cardiomyocyte proliferation and improved cardiac functions following myocardial infarction in adult mice, suggesting a potential method to enhance cardiac regeneration and improve cardiac function in the injured heart. Conclusions: This study demonstrates that CM-S1PR1 plays an essential role in cardiomyocyte proliferation and heart regeneration. This research provides a potential strategy by CM-targeted S1PR1 overexpression as a new therapeutic intervention for heart failure.
Collapse
Affiliation(s)
- Xiuxiang Liu
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Jinnan Yue
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Caixia Zhou
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Yunhao Duan
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Xiaoli Chen
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Jie Liu
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Shougang Zhuang
- Department of Medicine, Rhode Island Hospital and Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Yu Luo
- Department of Cardiology, Zigong Fourth People's Hospital, Sichuan, 643099, China
| | - Jinjin Wu
- Department of Cardiology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuzhen Zhang
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Lin Zhang
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Clinical Center for Heart Disease Research, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
6
|
Li G, Gao J, Ding P, Gao Y. The role of endothelial cell-pericyte interactions in vascularization and diseases. J Adv Res 2025; 67:269-288. [PMID: 38246244 PMCID: PMC11725166 DOI: 10.1016/j.jare.2024.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND Endothelial cells (ECs) and pericytes (PCs) are crucial components of the vascular system, with ECs lining the inner layer of blood vessels and PCs surrounding capillaries to regulate blood flow and angiogenesis. Intercellular communication between ECs and PCs is vital for the formation, stability, and function of blood vessels. Various signaling pathways, such as the vascular endothelial growth factor/vascular endothelial growth factor receptor pathway and the platelet-derived growth factor-B/platelet-derived growth factor receptor-β pathway, play roles in communication between ECs and PCs. Dysfunctional communication between these cells is associated with various diseases, including vascular diseases, central nervous system disorders, and certain types of cancers. AIM OF REVIEW This review aimed to explore the diverse roles of ECs and PCs in the formation and reshaping of blood vessels. This review focused on the essential signaling pathways that facilitate communication between these cells and investigated how disruptions in these pathways may contribute to disease. Additionally, the review explored potential therapeutic targets, future research directions, and innovative approaches, such as investigating the impact of EC-PCs in novel systemic diseases, addressing resistance to antiangiogenic drugs, and developing novel antiangiogenic medications to enhance therapeutic efficacy. KEY SCIENTIFIC CONCEPTS OF REVIEW Disordered EC-PC intercellular signaling plays a role in abnormal blood vessel formation, thus contributing to the progression of various diseases and the development of resistance to antiangiogenic drugs. Therefore, studies on EC-PC intercellular interactions have high clinical relevance.
Collapse
Affiliation(s)
- Gan Li
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Junjie Gao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; Shanghai Sixth People's Hospital Fujian, No. 16, Luoshan Section, Jinguang Road, Luoshan Street, Jinjiang City, Quanzhou, Fujian, China
| | - Peng Ding
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| | - Youshui Gao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| |
Collapse
|
7
|
Bass AJ, Cutler DJ, Epstein MP. A powerful framework for differential co-expression analysis of general risk factors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.29.626006. [PMID: 39677786 PMCID: PMC11642831 DOI: 10.1101/2024.11.29.626006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Differential co-expression analysis (DCA) aims to identify genes in a pathway whose shared expression depends on a risk factor. While DCA provides insights into the biological activity of diseases, existing methods are limited to categorical risk factors and/or suffer from bias due to batch and variance-specific effects. We propose a new framework, Kernel-based Differential Co-expression Analysis (KDCA), that harnesses correlation patterns between genes in a pathway to detect differential co-expression arising from general (i.e., continuous, discrete, or categorical) risk factors. Using various simulated pathway architectures, we find that KDCA accounts for common sources of bias to control the type I error rate while substantially increasing the power compared to the standard eigengene approach. We then applied KDCA to The Cancer Genome Atlas thyroid data set and found several differentially co-expressed pathways by age of diagnosis and BRAF mutation status that were undetected by the eigengene method. Collectively, our results demonstrate that KDCA is a powerful testing framework that expands DCA applications in expression studies.
Collapse
Affiliation(s)
- Andrew J. Bass
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - David J. Cutler
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK
| | | |
Collapse
|
8
|
Michael H, Weng GW, Vallas MM, Lovos D, Chen E, Sheiffele P, Weng W. Metabolomics analysis reveals resembling metabolites between humanized γδ TCR mice and human plasma. Sci Rep 2024; 14:29321. [PMID: 39592837 PMCID: PMC11599612 DOI: 10.1038/s41598-024-81003-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 11/22/2024] [Indexed: 11/28/2024] Open
Abstract
Gamma delta (γδ) T cells, which reside in mucosal and epithelial tissues, are integral to immune responses and are involved in various cancers, autoimmune, and infectious diseases. To study human γδ T cells to a translational level, we developed γδ humanized TCR-T1 (HuTCR-T1) mice using our TruHumanization platform. We compared the metabolomic profiles from plasma samples of wild-type (WT), γδ HuTCR-T1 mice, and humans using UHPLC-MS/MS. Untargeted metabolomics and lipidomics were used to screen all detectable metabolites. Principal component analysis revealed that the metabolomic profiles of γδ HuTCR-T1 mice closely resemble those of humans, with a clear segregation of metabolites between γδ HuTCR-T1 and WT mice. Most humanized γδ metabolites were classified as lipids, followed by organic compounds and amino acids. Pathway analysis identified significant alterations in the metabolism of tryptophan, tyrosine, sphingolipids, and glycerophospholipids, shifting these pathways towards a more human-like profile. Immunophenotyping showed that γδ HuTCR-T1 mice maintained normal proportions of both lymphoid and myeloid immune cell populations, closely resembling WT mice, with only a few exceptions. These findings demonstrate that the γδ HuTCR-T1 mouse model exhibits a metabolomic profile that is remarkably similar to that of humans, highlighting its potential as a relevant model for investigating the role of metabolites in disease development and progression. This model also offers an opportunity to discover therapeutic human TCRs.
Collapse
Affiliation(s)
- Husheem Michael
- InGenious Targeting Laboratory, 2200 Smithtown Avenue Ronkonkoma, Ronkonkoma, NY, 11779, United States of America.
| | - Gene W Weng
- InGenious Targeting Laboratory, 2200 Smithtown Avenue Ronkonkoma, Ronkonkoma, NY, 11779, United States of America
| | - Mikaela M Vallas
- InGenious Targeting Laboratory, 2200 Smithtown Avenue Ronkonkoma, Ronkonkoma, NY, 11779, United States of America
| | - Douglas Lovos
- InGenious Targeting Laboratory, 2200 Smithtown Avenue Ronkonkoma, Ronkonkoma, NY, 11779, United States of America
| | - Ellen Chen
- InGenious Targeting Laboratory, 2200 Smithtown Avenue Ronkonkoma, Ronkonkoma, NY, 11779, United States of America
| | - Paul Sheiffele
- InGenious Targeting Laboratory, 2200 Smithtown Avenue Ronkonkoma, Ronkonkoma, NY, 11779, United States of America
| | - Wei Weng
- InGenious Targeting Laboratory, 2200 Smithtown Avenue Ronkonkoma, Ronkonkoma, NY, 11779, United States of America.
| |
Collapse
|
9
|
Mogollón García HD, de Andrade Ferrazza R, Ochoa JC, de Athayde FF, Vidigal PMP, Wiltbank M, Kastelic JP, Sartori R, Ferreira JCP. Landscape transcriptomic analysis of bovine follicular cells during key phases of ovarian follicular development. Biol Res 2024; 57:76. [PMID: 39468655 PMCID: PMC11514973 DOI: 10.1186/s40659-024-00558-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 10/15/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND There are many gaps in our understanding of the mechanisms involved in ovarian follicular development in cattle, particularly regarding follicular deviation, acquisition of ovulatory capacity, and preovulatory changes. Molecular evaluations of ovarian follicular cells during follicular development in cattle, especially serial transcriptomic analyses across key growth phases, have not been reported. This study aims to address this gap by analyzing gene expression using RNA-seq in granulosa and antral cells recovered from ovarian follicular fluid during critical phases of ovarian follicular development in Holstein cows. RESULTS Integrated analysis of gene ontology (GO), gene set enrichment (GSEA), protein-protein interaction (PPI), and gene topology identified that differentially expressed genes (DEGs) in the largest ovarian follicles at deviation (Dev) were primarily involved in FSH-negative feedback, steroidogenesis, cell proliferation, apoptosis, and the prevention of early follicle rupture. In contrast, DEGs in the second largest follicles (DevF2) were mainly related to loss of cell viability, apoptosis, and immune cell invasion. In the dominant (PostDev) and preovulatory (PreOv) follicles, DEGs were associated with vascular changes and inflammatory responses. CONCLUSIONS The transcriptome of ovarian follicular fluid cells had a predominance of granulosa cells in the dominant follicle at deviation, with upregulation of genes involved in cell viability, steroidogenesis, and apoptosis prevention, whereas in the non-selected follicle there was upregulation of cell death-related transcripts. Immune cell transcripts increased significantly after deviation, particularly in preovulatory follicles, indicating strong intrafollicular chemotactic activity. We inferred that immune cell invasion occurred despite an intact basal lamina, contributing to follicular maturation.
Collapse
Affiliation(s)
- Henry David Mogollón García
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Rua Prof. Doutor Walter Mauricio Correa, s/n, Botucatu, São Paulo, 18618-681, Brazil
- Department of Genetic, Evolution, Microbiology and Immunology. Biology Institute, Campinas State University, Campinas, São Paulo, Brazil
- Computational Systems Biology Laboratory (CSBL), Institut Pasteur, University of São Paulo (USP), São Paulo, Brazil
| | | | - Julian Camilo Ochoa
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Rua Prof. Doutor Walter Mauricio Correa, s/n, Botucatu, São Paulo, 18618-681, Brazil
| | - Flávia Florencio de Athayde
- Department of Animal Production and Health, School of Veterinary Medicine, São Paulo State University (UNESP), Araçatuba, São Paulo, Brazil
| | | | - Milo Wiltbank
- Department of Animal & Dairy Sciences, University of Wisconsin-Madison, Madison, USA
| | | | - Roberto Sartori
- Department of Animal Science, Luiz de Queiroz College of Agriculture (ESALQ), University of São Paulo, Piracicaba, São Paulo, Brazil
| | - João Carlos Pinheiro Ferreira
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Rua Prof. Doutor Walter Mauricio Correa, s/n, Botucatu, São Paulo, 18618-681, Brazil.
| |
Collapse
|
10
|
Limbu KR, Chhetri RB, Kim S, Shrestha J, Oh YS, Baek DJ, Park EY. Targeting sphingosine 1-phosphate and sphingosine kinases in pancreatic cancer: mechanisms and therapeutic potential. Cancer Cell Int 2024; 24:353. [PMID: 39462385 PMCID: PMC11514880 DOI: 10.1186/s12935-024-03535-7] [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] [Received: 07/17/2024] [Accepted: 10/15/2024] [Indexed: 10/29/2024] Open
Abstract
Pancreatic cancer is known to be the most lethal cancer. Fewer new treatments are being developed for pancreatic cancer as compared to other cancers. The bioactive lipid S1P, which is mainly regulated by sphingosine kinase 1 (SK1) and sphingosine kinase 2 (SK2) enzymes, plays significant roles in pancreatic cancer initiation and exacerbation. S1P controls many signaling pathways to modulate the progression of pancreatic cancer through the G-coupled receptor S1PR1-5. Several papers reporting amelioration of pancreatic cancer via modulation of S1P levels or downstream signaling pathways have previously been published. In this paper, for the first time, we have reviewed the results of previous studies to understand how S1P and its receptors contribute to the development of pancreatic cancer, and whether S1P can be a therapeutic target. In addition, we have also reviewed papers dealing with the effects of SK1 and SK2, which are kinases that regulate the level of S1P, on the pathogenesis of pancreatic cancer. We have also listed available drugs that particularly focus on S1P, S1PRs, SK1, and SK2 for the treatment of pancreatic cancer. Through this review, we would like to suggest that the SK/S1P/S1PR signaling system can be an important target for treating pancreatic cancer, where a new treatment target is desperately warranted.
Collapse
Affiliation(s)
- Khem Raj Limbu
- College of Pharmacy, Mokpo National University, Joennam, 58554, South Korea
| | | | - Subin Kim
- College of Pharmacy, Mokpo National University, Joennam, 58554, South Korea
| | - Jitendra Shrestha
- Massachusetts General Hospital Cancer Center, Boston, MA, 02114, USA
| | - Yoon Sin Oh
- Department of Food and Nutrition, Eulji University, Seongnam, 13135, South Korea
| | - Dong Jae Baek
- College of Pharmacy, Mokpo National University, Joennam, 58554, South Korea.
| | - Eun-Young Park
- College of Pharmacy, Mokpo National University, Joennam, 58554, South Korea.
| |
Collapse
|
11
|
Foran D, Antoniades C, Akoumianakis I. Emerging Roles for Sphingolipids in Cardiometabolic Disease: A Rational Therapeutic Target? Nutrients 2024; 16:3296. [PMID: 39408263 PMCID: PMC11478599 DOI: 10.3390/nu16193296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/19/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Cardiovascular disease is a leading cause of morbidity and mortality. New research elucidates increasingly complex relationships between cardiac and metabolic health, giving rise to new possible therapeutic targets. Sphingolipids are a heterogeneous class of bioactive lipids with critical roles in normal human physiology. They have also been shown to play both protective and deleterious roles in the pathogenesis of cardiovascular disease. Ceramides are implicated in dysregulating insulin signalling, vascular endothelial function, inflammation, oxidative stress, and lipoprotein aggregation, thereby promoting atherosclerosis and vascular disease. Ceramides also advance myocardial disease by enhancing pathological cardiac remodelling and cardiomyocyte death. Glucosylceramides similarly contribute to insulin resistance and vascular inflammation, thus playing a role in atherogenesis and cardiometabolic dysfunction. Sphingosing-1-phosphate, on the other hand, may ameliorate some of the pathological functions of ceramide by protecting endothelial barrier integrity and promoting cell survival. Sphingosine-1-phosphate is, however, implicated in the development of cardiac fibrosis. This review will explore the roles of sphingolipids in vascular, cardiac, and metabolic pathologies and will evaluate the therapeutic potential in targeting sphingolipids with the aim of prevention and reversal of cardiovascular disease in order to improve long-term cardiovascular outcomes.
Collapse
Affiliation(s)
| | | | - Ioannis Akoumianakis
- Cardiovascular Medicine Division, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK; (D.F.); (C.A.)
| |
Collapse
|
12
|
Jia W, Yuan J, Zhang J, Li S, Lin W, Cheng B. Bioactive sphingolipids as emerging targets for signal transduction in cancer development. Biochim Biophys Acta Rev Cancer 2024; 1879:189176. [PMID: 39233263 DOI: 10.1016/j.bbcan.2024.189176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/06/2024]
Abstract
Sphingolipids, crucial components of cellular membranes, play a vital role in maintaining cellular structure and signaling integrity. Disruptions in sphingolipid metabolism are increasingly implicated in cancer development. Key bioactive sphingolipids, such as ceramides, sphingosine-1-phosphate (S1P), ceramide-1-phosphate (C1P), and glycosphingolipids, profoundly impact tumor biology. They influence the behavior of tumor cells, stromal cells, and immune cells, affecting tumor aggressiveness, angiogenesis, immune modulation, and extracellular matrix remodeling. Furthermore, abnormal expression of sphingolipids and their metabolizing enzymes modulates the secretion of tumor-derived extracellular vesicles (TDEs), which are key players in creating an immunosuppressive tumor microenvironment, remodeling the extracellular matrix, and facilitating oncogenic signaling within in situ tumors and distant pre-metastatic niches (PMNs). Understanding the role of sphingolipids in the biogenesis of tumor-derived extracellular vesicles (TDEs) and their bioactive contents can pave the way for new biomarkers in cancer diagnosis and prognosis, ultimately enhancing comprehensive tumor treatment strategies.
Collapse
Affiliation(s)
- Wentao Jia
- Department of General Practice, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; Oncology Department of Traditional Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; Faculty of Traditional Chinese Medicine, Naval Medical University, Shanghai 200043, China
| | - Jiaying Yuan
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Jinbo Zhang
- Department of Pharmacy, Tianjin Rehabilitation and Recuperation Center, Joint Logistics Support Force, Tianjin 300000, China
| | - Shu Li
- Department of Gastroenterology, Baoshan Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201900, China
| | - Wanfu Lin
- Oncology Department of Traditional Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; Faculty of Traditional Chinese Medicine, Naval Medical University, Shanghai 200043, China.
| | - Binbin Cheng
- Oncology Department of Traditional Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; Faculty of Traditional Chinese Medicine, Naval Medical University, Shanghai 200043, China.
| |
Collapse
|
13
|
Banerjee R, Knauer LA, Iyer D, Barlow SE, Shalaby H, Dehghan R, Scallan JP, Yang Y. Rictor, an mTORC2 Protein, Regulates Murine Lymphatic Valve Formation Through the AKT-FOXO1 Signaling. Arterioscler Thromb Vasc Biol 2024; 44:2004-2023. [PMID: 39087350 PMCID: PMC11335088 DOI: 10.1161/atvbaha.124.321164] [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: 04/29/2024] [Accepted: 07/10/2024] [Indexed: 08/02/2024]
Abstract
BACKGROUND Lymphatic valves are specialized structures in collecting lymphatic vessels and are crucial for preventing retrograde lymph flow. Mutations in valve-forming genes have been clinically implicated in the pathology of congenital lymphedema. Lymphatic valves form when oscillatory shear stress from lymph flow signals through the PI3K/AKT pathway to promote the transcription of valve-forming genes that trigger the growth and maintenance of lymphatic valves. Conventionally, in many cell types, AKT is phosphorylated at Ser473 by the mTORC2 (mammalian target of rapamycin complex 2). However, mTORC2 has not yet been implicated in lymphatic valve formation. METHODS In vivo and in vitro techniques were used to investigate the role of Rictor, a critical component of mTORC2, in lymphatic endothelium. RESULTS Here, we showed that embryonic and postnatal lymphatic deletion of Rictor, a critical component of mTORC2, led to a significant decrease in lymphatic valves and prevented the maturation of collecting lymphatic vessels. RICTOR knockdown in human dermal lymphatic endothelial cells not only reduced the level of activated AKT and the expression of valve-forming genes under no-flow conditions but also abolished the upregulation of AKT activity and valve-forming genes in response to oscillatory shear stress. We further showed that the AKT target, FOXO1 (forkhead box protein O1), a repressor of lymphatic valve formation, had increased nuclear activity in Rictor knockout mesenteric lymphatic endothelial cells in vivo. Deletion of Foxo1 in Rictor knockout mice restored the number of valves to control levels in lymphatic vessels of the ear and mesentery. CONCLUSIONS Our work identifies a novel role for RICTOR in the mechanotransduction signaling pathway, wherein it activates AKT and prevents the nuclear accumulation of the valve repressor, FOXO1, which ultimately enables the formation and maintenance of lymphatic valves.
Collapse
Affiliation(s)
- Richa Banerjee
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| | - Luz A. Knauer
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| | - Drishya Iyer
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| | - Sara E. Barlow
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| | - Hanan Shalaby
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| | - Razieh Dehghan
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| | - Joshua P. Scallan
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| | - Ying Yang
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| |
Collapse
|
14
|
Modak A, Mishra SR, Awasthi M, Aravind A, Singh S, Sreekumar E. Fingolimod (FTY720), an FDA-approved sphingosine 1-phosphate (S1P) receptor agonist, restores endothelial hyperpermeability in cellular and animal models of dengue virus serotype 2 infection. IUBMB Life 2024; 76:267-285. [PMID: 38031996 DOI: 10.1002/iub.2795] [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: 04/07/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023]
Abstract
Extensive vascular leakage and shock is a major cause of dengue-associated mortality. At present, there are no specific treatments available. Sphingolipid pathway is a key player in the endothelial barrier integrity; and is mediated through the five sphingosine-1-phosphate receptors (S1PR1-S1PR5). Signaling through S1PR2 promotes barrier disruption; and in Dengue virus (DENV)-infection, there is overexpression of this receptor. Fingolimod (FTY720) is a specific agonist that targets the remaining barrier-protective S1P receptors, without targeting S1PR2. In the present study, we explored whether FTY720 treatment can alleviate DENV-induced endothelial hyperpermeability. In functional assays, in both in vitro systems and in AG129 animal models, FTY720 treatment was found effective. Upon treatment, there was complete restoration of the monolayer integrity in DENV serotype 2-infected human microvascular endothelial cells (HMEC-1). At the molecular level, the treatment reversed activation of the S1P pathway. It significantly reduced the phosphorylation of the key molecules such as PTEN, RhoA, and VE-Cadherin; and also, the expression levels of S1PR2. In DENV2-infected AG129 mice treated with FTY720, there was significant improvement in weight gain, in overall clinical symptoms, and in survival. Whereas 100% of the DENV2-infected, untreated animals died by day-10 post-infection, 70% of the FTY720-treated animals were alive; and at the end of the 15-day post-infection observation period, 30% of them were still surviving. There was a significant reduction in the Evan's-blue dye permeability in the organs of FTY720-treated, DENV-2 infected animals; and also improvement in the hemogram, with complete restoration of thrombocytopenia and hepatic function. Our results show that the FDA-approved molecule Fingolimod (FTY720) is a promising therapeutic intervention in severe dengue.
Collapse
Affiliation(s)
- Ayan Modak
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, India
- Regional Centre for Biotechnology (RCB), NCR Biotech Science Cluster, Faridabad, Haryana (NCR Delhi), India
| | - Srishti Rajkumar Mishra
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, India
- Regional Centre for Biotechnology (RCB), NCR Biotech Science Cluster, Faridabad, Haryana (NCR Delhi), India
| | - Mansi Awasthi
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, India
- Regional Centre for Biotechnology (RCB), NCR Biotech Science Cluster, Faridabad, Haryana (NCR Delhi), India
| | - Arya Aravind
- Animal Research Facility, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, Kerala, India
| | - Sneha Singh
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, India
| | - Easwaran Sreekumar
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, India
- Molecular Bioassay Laboratory, Institute of Advanced Virology (IAV), Thiruvananthapuram, Kerala, India
| |
Collapse
|
15
|
Nagahashi M, Miyoshi Y. Targeting Sphingosine-1-Phosphate Signaling in Breast Cancer. Int J Mol Sci 2024; 25:3354. [PMID: 38542328 PMCID: PMC10970081 DOI: 10.3390/ijms25063354] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 03/06/2024] [Accepted: 03/12/2024] [Indexed: 01/04/2025] Open
Abstract
In recent years, newly emerging therapies, such as immune checkpoint inhibitors and antibody-drug conjugates, have further improved outcomes for breast cancer patients. However, recurrent and metastatic breast cancer often eventually develops resistance to these drugs, and cure is still rare. As such, the development of new therapies for refractory breast cancer that differ from conventional mechanisms of action is necessary. Sphingosine-1-phosphate (S1P) is a key molecule with a variety of bioactive activities, including involvement in cancer cell proliferation, invasion, and metastasis. S1P also contributes to the formation of the cancer microenvironment by inducing surrounding vascular- and lymph-angiogenesis and regulating the immune system. In this article, we outline the basic mechanism of action of S1P, summarize previous findings on the function of S1P in cancer cells and the cancer microenvironment, and discuss the clinical significance of S1P in breast cancer and the therapeutic potential of targeting S1P signaling.
Collapse
Affiliation(s)
- Masayuki Nagahashi
- Department of Surgery, Division of Breast and Endocrine Surgery, School of Medicine, Hyogo Medical University, 1-1 Mukogawa-cho, Nishinomiya 663-8501, Hyogo, Japan;
| | | |
Collapse
|
16
|
Sasazaki S, Kondo H, Moriishi Y, Kawaguchi F, Oyama K, Mannen H. Comprehensive genotyping analysis of single nucleotide polymorphisms responsible for beef marbling in Japanese Black cattle. BMC Genom Data 2024; 25:17. [PMID: 38336623 PMCID: PMC10854043 DOI: 10.1186/s12863-024-01199-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND Beef marbling is considered a desirable trait in the meat industry. Therefore, understanding the genetic factors that cause marbling is important. Previously, we performed a genome-wide association study to examine genetic factors associated with beef marbling in Japanese Black cattle and identified a candidate region between 10-30 Mbp on chromosome 7. We verified the effect of the SNPs in this region on beef marbling using linkage disequilibrium block analysis. We narrowed down the candidate region to a range of 15.8-16.1 Mbp. In this study, we comprehensively detected all of the SNPs in this region and verified their effects on beef marbling. RESULTS Genome resequencing using four animals exhibiting high beef marbling standard (BMS) and four with low BMS revealed a total of 1,846 polymorphisms within the candidate region. Based on the annotation, we selected 13 SNPs exhibiting a moderate impact, as no high-impact SNPs were detected. All of the SNPs represented missense polymorphisms and were located in the following seven genes: RDH8, ANGPTL6, DNMT1, MRPL4, ICAM1, ICAM3, and ICAM5. Finally, we determined the effects of these SNPs on the BMS of a Japanese Black cattle population (n = 529). Analysis of variance revealed that the five SNPs were located in genes encoding the intercellular adhesion molecules (ICAM1, ICAM3, and ICAM5), and showed a highly significant association compared with the remainder (p < 0.01). The lowest p-value was observed for ICAM3_c.739G > A (p = 1.18E-04). Previous studies have suggested that intercellular adhesion molecules (ICAM) may be an upstream factor that regulates adipocyte differentiation. Therefore, considering the polymorphism and putative gene function, we suggest that ICAM1 is potentially responsible for beef marbling. c.470C > G and/or c.994G > A on ICAM1 may be responsible for this quantitative trait locus. CONCLUSIONS Promising SNP candidates responsible for beef marbling were identified using extensive polymorphism verification in a previously reported QTL region. We aim to elucidate the mechanism of beef marbling in future studies by investigating how these polymorphisms alter protein structure and function.
Collapse
Affiliation(s)
- Shinji Sasazaki
- Laboratory of Animal Breeding and Genetics, Graduate School of Agricultural Science, Kobe University, Kobe, Japan.
| | - Hina Kondo
- Laboratory of Animal Breeding and Genetics, Graduate School of Agricultural Science, Kobe University, Kobe, Japan
| | - Yurika Moriishi
- Laboratory of Animal Breeding and Genetics, Graduate School of Agricultural Science, Kobe University, Kobe, Japan
| | - Fuki Kawaguchi
- Laboratory of Animal Breeding and Genetics, Graduate School of Agricultural Science, Kobe University, Kobe, Japan
| | - Kenji Oyama
- Food Resources Education & Research Center, Kobe University, Kasai, Japan
| | - Hideyuki Mannen
- Laboratory of Animal Breeding and Genetics, Graduate School of Agricultural Science, Kobe University, Kobe, Japan
| |
Collapse
|
17
|
Xu T, Jiang Y, Fu H, Yang G, Hu X, Chen Y, Zhang Q, Wang Y, Wang Y, Xie HQ, Han F, Xu L, Zhao B. Exploring the adverse effects of 1,3,6,8-tetrabromo-9H-carbazole in atherosclerotic model mice by metabolomic profiling integrated with mechanism studies in vitro. CHEMOSPHERE 2024; 349:140767. [PMID: 37992903 DOI: 10.1016/j.chemosphere.2023.140767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 10/04/2023] [Accepted: 11/17/2023] [Indexed: 11/24/2023]
Abstract
Given its wide distribution in the environment and latent toxic effects, 1,3,6,8-tetrabromo-9H-carbazole (1368-BCZ) is an emerging concern that has gained increasing attention globally. 1368-BCZ exposure is reported to have potential cardiovascular toxicity. Although atherosclerosis is a cardiovascular disease and remains a primary cause of mortality worldwide, no evidence has been found regarding the impact of 1368-BCZ on atherosclerosis. Therefore, we aimed to explore the deleterious effects of 1368-BCZ on atherosclerosis and the underlying mechanisms. Serum samples from 1368-BCZ-treated atherosclerotic model mice were subjected to metabolomic profiling to investigate the adverse influence of the pollutant. Subsequently, the molecular mechanism associated with the metabolic pathway of atherosclerotic mice that was identified following 1368-BCZ exposure was validated in vitro. Serum metabolomics analysis revealed that 1368-BCZ significantly altered the tricarboxylic acid cycle, causing a disturbance in energy metabolism. In vitro, we further validated general markers of energy metabolism based on metabolome data: 1368-BCZ dampened adenosine triphosphate (ATP) synthesis and increased reactive oxygen species (ROS) production. Furthermore, blocking the aryl hydrocarbon receptor (AhR) reversed the high production of ROS induced by 1368-BCZ. It is concluded that 1368-BCZ decreased the ATP synthesis by disturbing the energy metabolism, thereby stimulating the AhR-mediated ROS production and presumably causing aggravated atherosclerosis. This is the first comprehensive study on the cardiovascular toxicity and mechanism of 1368-BCZ based on rodent models of atherosclerosis and integrated with in vitro models.
Collapse
Affiliation(s)
- Tong Xu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; University of Chinese Academy of Sciences, Beijing, 100049, China; PET/CT Center, Key Laboratory of Functional Molecular Imaging, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, China
| | - Yu Jiang
- Department of Preventive Medicine, Fujian Provincial Key Laboratory of Environment Factors and Cancer, Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, 350122, China
| | - Hualing Fu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Guanglei Yang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaoxu Hu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yangsheng Chen
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qian Zhang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Beijing, 100730, China
| | - Yuxi Wang
- Department of Preventive Medicine, Fujian Provincial Key Laboratory of Environment Factors and Cancer, Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, 350122, China
| | - Yilan Wang
- PET/CT Center, Key Laboratory of Functional Molecular Imaging, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, China
| | - Heidi Qunhui Xie
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fang Han
- PET/CT Center, Key Laboratory of Functional Molecular Imaging, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, China.
| | - Li Xu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Bin Zhao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
18
|
Liu J, Liu X, Luo Y, Huang F, Xie Y, Zheng S, Jia B, Xiao Z. Sphingolipids: drivers of cardiac fibrosis and atrial fibrillation. J Mol Med (Berl) 2024; 102:149-165. [PMID: 38015241 PMCID: PMC10858135 DOI: 10.1007/s00109-023-02391-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 10/13/2023] [Accepted: 10/23/2023] [Indexed: 11/29/2023]
Abstract
Sphingolipids (SLs) are vital constituents of the plasma membrane of animal cells and concurrently regulate numerous cellular processes. An escalating number of research have evinced that SLs assume a crucial part in the progression of tissue fibrosis, a condition for which no efficacious cure exists as of now. Cardiac fibrosis, and in particular, atrial fibrosis, is a key factor in the emergence of atrial fibrillation (AF). AF has become one of the most widespread cardiac arrhythmias globally, with its incidence continuing to mount, thereby propelling it to the status of a major public health concern. This review expounds on the structure and biosynthesis pathways of several pivotal SLs, the pathophysiological mechanisms of AF, and the function of SLs in cardiac fibrosis. Delving into the influence of sphingolipid levels in the alleviation of cardiac fibrosis offers innovative therapeutic strategies to address cardiac fibrosis and AF.
Collapse
Affiliation(s)
- Junjie Liu
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ximao Liu
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yucheng Luo
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fangze Huang
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yu Xie
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shaoyi Zheng
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Bo Jia
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China.
| | - Zezhou Xiao
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
19
|
Liu XT, Huang Y, Liu D, Jiang YC, Zhao M, Chung LH, Han XD, Zhao Y, Chen J, Coleman P, Ting KK, Tran C, Su Y, Dennis CV, Bhatnagar A, Liu K, Don AS, Vadas MA, Gorrell MD, Zhang S, Murray M, Kavurma MM, McCaughan GW, Gamble JR, Qi Y. Targeting the SphK1/S1P/PFKFB3 axis suppresses hepatocellular carcinoma progression by disrupting glycolytic energy supply that drives tumor angiogenesis. J Transl Med 2024; 22:43. [PMID: 38200582 PMCID: PMC10782643 DOI: 10.1186/s12967-023-04830-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 12/24/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) remains a leading life-threatening health challenge worldwide, with pressing needs for novel therapeutic strategies. Sphingosine kinase 1 (SphK1), a well-established pro-cancer enzyme, is aberrantly overexpressed in a multitude of malignancies, including HCC. Our previous research has shown that genetic ablation of Sphk1 mitigates HCC progression in mice. Therefore, the development of PF-543, a highly selective SphK1 inhibitor, opens a new avenue for HCC treatment. However, the anti-cancer efficacy of PF-543 has not yet been investigated in primary cancer models in vivo, thereby limiting its further translation. METHODS Building upon the identification of the active form of SphK1 as a viable therapeutic target in human HCC specimens, we assessed the capacity of PF-543 in suppressing tumor progression using a diethylnitrosamine-induced mouse model of primary HCC. We further delineated its underlying mechanisms in both HCC and endothelial cells. Key findings were validated in Sphk1 knockout mice and lentiviral-mediated SphK1 knockdown cells. RESULTS SphK1 activity was found to be elevated in human HCC tissues. Administration of PF-543 effectively abrogated hepatic SphK1 activity and significantly suppressed HCC progression in diethylnitrosamine-treated mice. The primary mechanism of action was through the inhibition of tumor neovascularization, as PF-543 disrupted endothelial cell angiogenesis even in a pro-angiogenic milieu. Mechanistically, PF-543 induced proteasomal degradation of the critical glycolytic enzyme 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3, thus restricting the energy supply essential for tumor angiogenesis. These effects of PF-543 could be reversed upon S1P supplementation in an S1P receptor-dependent manner. CONCLUSIONS This study provides the first in vivo evidence supporting the potential of PF-543 as an effective anti-HCC agent. It also uncovers previously undescribed links between the pro-cancer, pro-angiogenic and pro-glycolytic roles of the SphK1/S1P/S1P receptor axis. Importantly, unlike conventional anti-HCC drugs that target individual pro-angiogenic drivers, PF-543 impairs the PFKFB3-dictated glycolytic energy engine that fuels tumor angiogenesis, representing a novel and potentially safer therapeutic strategy for HCC.
Collapse
Affiliation(s)
- Xin Tracy Liu
- Centenary Institute of Cancer Medicine and Cell Biology, The University of Sydney, Sydney, NSW, 2050, Australia
| | - Yu Huang
- Centenary Institute of Cancer Medicine and Cell Biology, The University of Sydney, Sydney, NSW, 2050, Australia
| | - Da Liu
- Centenary Institute of Cancer Medicine and Cell Biology, The University of Sydney, Sydney, NSW, 2050, Australia
| | - Yingxin Celia Jiang
- Centenary Institute of Cancer Medicine and Cell Biology, The University of Sydney, Sydney, NSW, 2050, Australia
| | - Min Zhao
- School of Science, Technology and Engineering, University of the Sunshine Coast, Maroochydore DC, QLD, 4558, Australia
| | - Long Hoa Chung
- Centenary Institute of Cancer Medicine and Cell Biology, The University of Sydney, Sydney, NSW, 2050, Australia
| | - Xingxing Daisy Han
- Centenary Institute of Cancer Medicine and Cell Biology, The University of Sydney, Sydney, NSW, 2050, Australia
| | - Yinan Zhao
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, Dalian Minzu University, Dalian, 116600, Liaoning, China
| | - Jinbiao Chen
- Centenary Institute of Cancer Medicine and Cell Biology, The University of Sydney, Sydney, NSW, 2050, Australia
| | - Paul Coleman
- Centenary Institute of Cancer Medicine and Cell Biology, The University of Sydney, Sydney, NSW, 2050, Australia
| | - Ka Ka Ting
- Centenary Institute of Cancer Medicine and Cell Biology, The University of Sydney, Sydney, NSW, 2050, Australia
| | - Collin Tran
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Yingying Su
- Sydney Microscopy and Microanalysis, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Claude Vincent Dennis
- AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Sydney Local Health District, Sydney, NSW, 2050, Australia
| | - Atul Bhatnagar
- Sydney Mass Spectrometry, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Ken Liu
- AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Sydney Local Health District, Sydney, NSW, 2050, Australia
| | - Anthony Simon Don
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Mathew Alexander Vadas
- Centenary Institute of Cancer Medicine and Cell Biology, The University of Sydney, Sydney, NSW, 2050, Australia
| | - Mark Douglas Gorrell
- Centenary Institute of Cancer Medicine and Cell Biology, The University of Sydney, Sydney, NSW, 2050, Australia
- AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Sydney Local Health District, Sydney, NSW, 2050, Australia
| | - Shubiao Zhang
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, Dalian Minzu University, Dalian, 116600, Liaoning, China
| | - Michael Murray
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia
| | | | - Geoffrey William McCaughan
- Centenary Institute of Cancer Medicine and Cell Biology, The University of Sydney, Sydney, NSW, 2050, Australia
- AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Sydney Local Health District, Sydney, NSW, 2050, Australia
| | - Jennifer Ruth Gamble
- Centenary Institute of Cancer Medicine and Cell Biology, The University of Sydney, Sydney, NSW, 2050, Australia
| | - Yanfei Qi
- Centenary Institute of Cancer Medicine and Cell Biology, The University of Sydney, Sydney, NSW, 2050, Australia.
| |
Collapse
|
20
|
Xu X, Han Y, Zhu T, Fan F, Wang X, Liu Y, Luo D. The role of SphK/S1P/S1PR signaling pathway in bone metabolism. Biomed Pharmacother 2023; 169:115838. [PMID: 37944444 DOI: 10.1016/j.biopha.2023.115838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/27/2023] [Accepted: 11/01/2023] [Indexed: 11/12/2023] Open
Abstract
There are a large number of people worldwide who suffer from osteoporosis, which imposes a huge economic burden, so it is necessary to explore the underlying mechanisms to achieve better supportive and curative care outcomes. Sphingosine kinase (SphK) is an enzyme that plays a crucial role in the synthesis of sphingosine-1-phosphate (S1P). S1P with paracrine and autocrine activities that act through its cell surface S1P receptors (S1PRs) and intracellular signals. In osteoporosis, S1P is indispensable for both normal and disease conditions. S1P has complicated roles in regulating osteoblast and osteoclast, respectively, and there have been exciting developments in understanding how SphK/S1P/S1PR signaling regulates these processes in response to osteoporosis therapy. Here, we review the proliferation, differentiation, apoptosis, and functions of S1P, specifically detailing the roles of S1P and S1PRs in osteoblasts and osteoclasts. Finally, we focus on the S1P-based therapeutic approaches in bone metabolism, which may provide valuable insights into potential therapeutic strategies for osteoporosis.
Collapse
Affiliation(s)
- Xuefeng Xu
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, China
| | - Yi Han
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, China
| | - Tianxin Zhu
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, China
| | - Faxin Fan
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, China
| | - Xin Wang
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, China
| | - Yuqing Liu
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, China
| | - Duosheng Luo
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, China.
| |
Collapse
|
21
|
Xiao J. Sphingosine 1-Phosphate Lyase in the Developing and Injured Nervous System: a Dichotomy? Mol Neurobiol 2023; 60:6869-6882. [PMID: 37507574 PMCID: PMC10657793 DOI: 10.1007/s12035-023-03524-3] [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: 04/12/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023]
Abstract
Sphingosine 1-phosphate lyase (SPL) is the terminal enzyme that controls the degradation of the bioactive lipid sphingosine 1-phosphate (S1P) within an interconnected sphingolipid metabolic network. The unique metabolic position of SPL in maintaining S1P levels implies SPL could be an emerging new therapeutic target. Over the past decade, an evolving effort has been made to unravel the role of SPL in the nervous system; however, to what extent SPL influences the developing and mature nervous system through altering S1P biosynthesis remains opaque. While congenital SPL deletion is associated with deficits in the developing nervous system, the loss of SPL activity in adults appears to be neuroprotective in acquired neurological disorders. The controversial findings concerning SPL's role in the nervous system are further constrained by the current genetic and pharmacological tools. This review attempts to focus on the multi-faceted nature of SPL function in the mammalian nervous systems, implying its dichotomy in the developing and adult central nervous system (CNS). This article also highlights SPL is emerging as a therapeutic molecule that can be selectively targeted to modulate S1P for the treatment of acquired neurodegenerative diseases, raising new questions for future investigation. The development of cell-specific inducible conditional SPL mutants and selective pharmacological tools will allow the precise understanding of SPL's function in the adult CNS, which will aid the development of a new strategy focusing on S1P-based therapies for neuroprotection.
Collapse
Affiliation(s)
- Junhua Xiao
- Department of Health Sciences and Biostatistics, School of Health Sciences, Swinburne University of Technology, John Street, Hawthorn, VIC, 3022, Australia.
| |
Collapse
|
22
|
Li J, Huang Y, Zhang Y, Liu P, Liu M, Zhang M, Wu R. S1P/S1PR signaling pathway advancements in autoimmune diseases. BIOMOLECULES & BIOMEDICINE 2023; 23:922-935. [PMID: 37504219 PMCID: PMC10655875 DOI: 10.17305/bb.2023.9082] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 07/06/2023] [Accepted: 07/06/2023] [Indexed: 07/29/2023]
Abstract
Sphingosine-1-phosphate (S1P) is a versatile sphingolipid that is generated through the phosphorylation of sphingosine by sphingosine kinase (SPHK). S1P exerts its functional effects by binding to the G protein-coupled S1P receptor (S1PR). This lipid mediator plays a pivotal role in various cellular activities. The S1P/S1PR signaling pathway is implicated in the pathogenesis of immune-mediated diseases, significantly contributing to the functioning of the immune system. It plays a crucial role in diverse physiological and pathophysiological processes, including cell survival, proliferation, migration, immune cell recruitment, synthesis of inflammatory mediators, and the formation of lymphatic and blood vessels. However, the full extent of the involvement of this signaling pathway in the development of autoimmune diseases remains to be fully elucidated. Therefore, this study aims to comprehensively review recent research on the S1P/S1PR axis in diseases related to autoimmunity.
Collapse
Affiliation(s)
- Jianbin Li
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yiping Huang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yueqin Zhang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Pengcheng Liu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Mengxia Liu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Min Zhang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Rui Wu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
23
|
Torcasio R, Gallo Cantafio ME, Ikeda RK, Ganino L, Viglietto G, Amodio N. Lipid metabolic vulnerabilities of multiple myeloma. Clin Exp Med 2023; 23:3373-3390. [PMID: 37639069 PMCID: PMC10618328 DOI: 10.1007/s10238-023-01174-2] [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: 07/13/2023] [Accepted: 08/15/2023] [Indexed: 08/29/2023]
Abstract
Multiple myeloma (MM) is the second most common hematological malignancy worldwide, characterized by abnormal proliferation of malignant plasma cells within a tumor-permissive bone marrow microenvironment. Metabolic dysfunctions are emerging as key determinants in the pathobiology of MM. In this review, we highlight the metabolic features of MM, showing how alterations in various lipid pathways, mainly involving fatty acids, cholesterol and sphingolipids, affect the growth, survival and drug responsiveness of MM cells, as well as their cross-talk with other cellular components of the tumor microenvironment. These findings will provide a new path to understanding the mechanisms underlying how lipid vulnerabilities may arise and affect the phenotype of malignant plasma cells, highlighting novel druggable pathways with a significant impact on the management of MM.
Collapse
Affiliation(s)
- Roberta Torcasio
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Viale Europa, Campus Germaneto, 88100, Catanzaro, Italy
- Department of Biology, Ecology and Heart Sciences, University of Calabria, Arcavacata Di Rende, Cosenza, Italy
| | - Maria Eugenia Gallo Cantafio
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Viale Europa, Campus Germaneto, 88100, Catanzaro, Italy
| | - Raissa Kaori Ikeda
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Viale Europa, Campus Germaneto, 88100, Catanzaro, Italy
- Centro Universitário São Camilo, São Paulo, Brazil
| | - Ludovica Ganino
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Viale Europa, Campus Germaneto, 88100, Catanzaro, Italy
| | - Giuseppe Viglietto
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Viale Europa, Campus Germaneto, 88100, Catanzaro, Italy
| | - Nicola Amodio
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Viale Europa, Campus Germaneto, 88100, Catanzaro, Italy.
| |
Collapse
|
24
|
Sun G, Wang B, Zhu H, Ye J, Liu X. Role of sphingosine 1-phosphate (S1P) in sepsis-associated intestinal injury. Front Med (Lausanne) 2023; 10:1265398. [PMID: 37746079 PMCID: PMC10514503 DOI: 10.3389/fmed.2023.1265398] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 08/28/2023] [Indexed: 09/26/2023] Open
Abstract
Sphingosine-1-phosphate (S1P) is a widespread lipid signaling molecule that binds to five sphingosine-1-phosphate receptors (S1PRs) to regulate downstream signaling pathways. Sepsis can cause intestinal injury and intestinal injury can aggravate sepsis. Thus, intestinal injury and sepsis are mutually interdependent. S1P is more abundant in intestinal tissues as compared to other tissues, exerts anti-inflammatory effects, promotes immune cell trafficking, and protects the intestinal barrier. Despite the clinical importance of S1P in inflammation, with a very well-defined mechanism in inflammatory bowel disease, their role in sepsis-induced intestinal injury has been relatively unexplored. In addition to regulating lymphocyte exit, the S1P-S1PR pathway has been implicated in the gut microbiota, intestinal epithelial cells (IECs), and immune cells in the lamina propria. This review mainly elaborates on the physiological role of S1P in sepsis, focusing on intestinal injury. We introduce the generation and metabolism of S1P, emphasize the maintenance of intestinal barrier homeostasis in sepsis, and the protective effect of S1P in the intestine. We also review the link between sepsis-induced intestinal injury and S1P-S1PRs signaling, as well as the underlying mechanisms of action. Finally, we discuss how S1PRs affect intestinal function and become targets for future drug development to improve the translational capacity of preclinical studies to the clinic.
Collapse
Affiliation(s)
- Gehui Sun
- Gannan Medical University, Ganzhou, Jiangxi, China
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Bin Wang
- Gannan Medical University, Ganzhou, Jiangxi, China
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Hongquan Zhu
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Critical Care Medicine, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Junming Ye
- Gannan Medical University, Ganzhou, Jiangxi, China
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Xiaofeng Liu
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
- Department of Emergency, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| |
Collapse
|
25
|
Yu X. Promising Therapeutic Treatments for Cardiac Fibrosis: Herbal Plants and Their Extracts. Cardiol Ther 2023; 12:415-443. [PMID: 37247171 PMCID: PMC10423196 DOI: 10.1007/s40119-023-00319-4] [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: 02/28/2023] [Accepted: 04/27/2023] [Indexed: 05/30/2023] Open
Abstract
Cardiac fibrosis is closely associated with multiple heart diseases, which are a prominent health issue in the global world. Neurohormones and cytokines play indispensable roles in cardiac fibrosis. Many signaling pathways participate in cardiac fibrosis as well. Cardiac fibrosis is due to impaired degradation of collagen and impaired fibroblast activation, and collagen accumulation results in increasing heart stiffness and inharmonious activity, leading to structure alterations and finally cardiac function decline. Herbal plants have been applied in traditional medicines for thousands of years. Because of their naturality, they have attracted much attention for use in resisting cardiac fibrosis in recent years. This review sheds light on several extracts from herbal plants, which are promising therapeutics for reversing cardiac fibrosis.
Collapse
Affiliation(s)
- Xuejing Yu
- Department of Internal Medicine, Division of Cardiology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75235, USA.
| |
Collapse
|
26
|
Hallisey VM, Schwab SR. Get me out of here: Sphingosine 1-phosphate signaling and T cell exit from tissues during an immune response. Immunol Rev 2023; 317:8-19. [PMID: 37212181 DOI: 10.1111/imr.13219] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 05/23/2023]
Abstract
During an immune response, the duration of T cell residence in lymphoid and non-lymphoid tissues likely affects T cell activation, differentiation, and memory development. The factors that govern T cell transit through inflamed tissues remain incompletely understood, but one important determinant of T cell exit from tissues is sphingosine 1-phosphate (S1P) signaling. In homeostasis, S1P levels are high in blood and lymph compared to lymphoid organs, and lymphocytes follow S1P gradients out of tissues into circulation using varying combinations of five G-protein coupled S1P receptors. During an immune response, both the shape of S1P gradients and the expression of S1P receptors are dynamically regulated. Here we review what is known, and key questions that remain unanswered, about how S1P signaling is regulated in inflammation and in turn how S1P shapes immune responses.
Collapse
Affiliation(s)
- Victoria M Hallisey
- Department of Cell Biology, New York University Grossman School of Medicine, New York, New York, USA
| | - Susan R Schwab
- Department of Cell Biology, New York University Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
27
|
Alam S, Afsar SY, Wolter MA, Volk LM, Mitroi DN, Meyer Zu Heringdorf D, van Echten-Deckert G. S1P Lyase Deficiency in the Brain Promotes Astrogliosis and NLRP3 Inflammasome Activation via Purinergic Signaling. Cells 2023; 12:1844. [PMID: 37508508 PMCID: PMC10378183 DOI: 10.3390/cells12141844] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/19/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
Astrocytes are critical players in brain health and disease. Brain pathologies and lesions are usually accompanied by astroglial alterations known as reactive astrogliosis. Sphingosine 1-phosphate lyase (SGPL1) catalysis, the final step in sphingolipid catabolism, irreversibly cleaves its substrate sphingosine 1-phosphate (S1P). We have shown that neural ablation of SGPL1 causes accumulation of S1P and hence neuronal damage, cognitive deficits, as well as microglial activation. Moreover, the S1P/S1P-receptor signaling axis enhances ATP production in SGPL1-deficient astrocytes. Using immunohistochemical methods as well as RNA Seq and CUT&Tag we show how S1P signaling causes activation of the astrocytic purinoreceptor P2Y1 (P2Y1R). With specific pharmacological agonists and antagonists, we uncover the P2Y1R as the key player in S1P-induced astrogliosis, and DDX3X mediated the activation of the NLRP3 inflammasome, including caspase-1 and henceforward generation of interleukin-1ß (IL-1ß) and of other proinflammatory cytokines. Our results provide a novel route connecting S1P metabolism and signaling with astrogliosis and the activation of the NLRP3 inflammasome, a central player in neuroinflammation, known to be crucial for the pathogenesis of numerous brain illnesses. Thus, our study opens the door for new therapeutic strategies surrounding S1P metabolism and signaling in the brain.
Collapse
Affiliation(s)
- Shah Alam
- LIMES Institute for Membrane Biology and Lipid Biochemistry, Kekulé-Institute, University of Bonn, 53115 Bonn, Germany
| | - Sumaiya Yasmeen Afsar
- LIMES Institute for Membrane Biology and Lipid Biochemistry, Kekulé-Institute, University of Bonn, 53115 Bonn, Germany
| | - Maya Anik Wolter
- LIMES Institute for Membrane Biology and Lipid Biochemistry, Kekulé-Institute, University of Bonn, 53115 Bonn, Germany
| | - Luisa Michelle Volk
- Institute for General Pharmacology and Toxicology, University Hospital, Goethe University Frankfurt am Main, 60590 Frankfurt am Main, Germany
| | - Daniel Nicolae Mitroi
- LIMES Institute for Membrane Biology and Lipid Biochemistry, Kekulé-Institute, University of Bonn, 53115 Bonn, Germany
| | - Dagmar Meyer Zu Heringdorf
- Institute for General Pharmacology and Toxicology, University Hospital, Goethe University Frankfurt am Main, 60590 Frankfurt am Main, Germany
| | - Gerhild van Echten-Deckert
- LIMES Institute for Membrane Biology and Lipid Biochemistry, Kekulé-Institute, University of Bonn, 53115 Bonn, Germany
| |
Collapse
|
28
|
Zhang F, Lu Y. The Sphingosine 1-Phosphate Axis: an Emerging Therapeutic Opportunity for Endometriosis. Reprod Sci 2023; 30:2040-2059. [PMID: 36662421 PMCID: PMC9857924 DOI: 10.1007/s43032-023-01167-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 01/04/2023] [Indexed: 01/21/2023]
Abstract
Endometriosis is a common condition in women of reproductive age, but its current interventions are unsatisfactory. Recent research discovered a dysregulation of the sphingosine 1-phosphate (S1P) signaling pathway in endometriosis and showed a positive outcome by targeting it. The S1P axis participates in a series of fundamental pathophysiological processes. This narrative review is trying to expound the reported and putative (due to limited reports in this area for now) interactions between the S1P axis and endometriosis in those pathophysiological processes, to provide some perspectives for future research. In short, S1P signaling pathway is highly activated in the endometriotic lesion. The S1P concentration has a surge in the endometriotic cyst fluid and the peritoneal fluid, with the downstream dysregulation of its receptors. The S1P axis plays an essential role in the migration and activation of the immune cells, fibrosis, angiogenesis, pain-related hyperalgesia, and innervation. S1P receptor (S1PR) modulators showed an impressive therapeutic effect by targeting the different S1P receptors in the endometriosis model, and many other conditions resemble endometriosis. And several of them already got approval for clinical application in many diseases, which means a drug repurposing direction and a rapid clinical translation for endometriosis treatments.
Collapse
Affiliation(s)
- Fengrui Zhang
- Department of Gynecology, The Obstetrics & Gynecology Hospital of Fudan University, 419 Fangxie Rd, Shanghai, 200011, People's Republic of China
| | - Yuan Lu
- Department of Gynecology, The Obstetrics & Gynecology Hospital of Fudan University, 419 Fangxie Rd, Shanghai, 200011, People's Republic of China.
| |
Collapse
|
29
|
Garabet W, Shabes P, Wolters KH, Rembe JD, Ibing W, Wagenhäuser MU, Simon F, Schelzig H, Oberhuber A. Effect of Gelatin-Based Hemostats on Fibroblasts and Relevant Growth Factors in Wound Healing. Gels 2023; 9:504. [PMID: 37367174 DOI: 10.3390/gels9060504] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 06/18/2023] [Accepted: 06/19/2023] [Indexed: 06/28/2023] Open
Abstract
Gelatin-based hemostats have been used in various surgical fields and showed advantageous effects on central aspects of wound healing when compared to cellulose-based hemostats. Nevertheless, the influence of gelatin-based hemostats on wound healing has not been fully explored yet. Hemostats were applied to fibroblast cell cultures for 5, 30, 60 min, 24 h, 7 and 14 days and measurements were taken at 3, 6, 12, 24 h and 7 or 14 days, respectively. Cell proliferation was quantified after different exposure times and a contraction assay was conducted to measure the extent of the extracellular matrix over time. We further assessed quantitative levels of vascular endothelial growth factor and basic fibroblast growth factor using enzyme-linked immunosorbent assay. Fibroblast counts decreased significantly at 7 and 14 days independent of the application duration (p < 0.001 for 5 min application). The gelatin-based hemostat did not have a negative impact on cell matrix contraction. After application of gelatin-based hemostat, the basic fibroblast growth factor did not change; yet, the vascular endothelial growth factor significantly increased after a prolonged 24 h application time when compared to controls or to a 6 h exposure (p < 0.05). Gelatin-based hemostats did not impair contraction of the extracellular matrix or growth factor production (vascular endothelial growth factor and basic fibroblast growth factor), while cell proliferation diminished at late time points. In conclusion, the gelatin-based material seems to be compatible with central aspects of wound healing. For further clinical assessment, future animal and human studies are necessary.
Collapse
Affiliation(s)
- Waseem Garabet
- Department of Vascular and Endovascular Surgery, University Hospital of Düsseldorf, 40225 Düsseldorf, Germany
| | - Polina Shabes
- Department of Vascular and Endovascular Surgery, University Hospital of Düsseldorf, 40225 Düsseldorf, Germany
| | - Katharina Henrika Wolters
- Department of Vascular and Endovascular Surgery, University Hospital of Düsseldorf, 40225 Düsseldorf, Germany
| | - Julian-Dario Rembe
- Department of Vascular and Endovascular Surgery, University Hospital of Düsseldorf, 40225 Düsseldorf, Germany
| | - Wiebke Ibing
- Department of Vascular and Endovascular Surgery, University Hospital of Düsseldorf, 40225 Düsseldorf, Germany
| | - Markus Udo Wagenhäuser
- Department of Vascular and Endovascular Surgery, University Hospital of Düsseldorf, 40225 Düsseldorf, Germany
| | - Florian Simon
- Department of Vascular and Endovascular Surgery, University Hospital of Düsseldorf, 40225 Düsseldorf, Germany
| | - Hubert Schelzig
- Department of Vascular and Endovascular Surgery, University Hospital of Düsseldorf, 40225 Düsseldorf, Germany
| | - Alexander Oberhuber
- Department of Vascular and Endovascular Surgery, University Hospital of Münster, 48149 Münster, Germany
| |
Collapse
|
30
|
Banerjee R, Knauer LA, Iyer D, Barlow SE, Scallan JP, Yang Y. Rictor induces AKT signaling to regulate lymphatic valve formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.12.544698. [PMID: 37397997 PMCID: PMC10312634 DOI: 10.1101/2023.06.12.544698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Lymphatic valves are specialized structures of the collecting lymphatic vessels and are crucial for preventing retrograde lymph flow. Mutations in valve-forming genes have been clinically implicated in the pathology of congenital lymphedema. Lymphatic valves form when oscillatory shear stress (OSS) from lymph flow signals through the PI3K/AKT pathway to promote the transcription of valve-forming genes that trigger the growth and maintenance of lymphatic valves throughout life. Conventionally, in other tissue types, AKT activation requires dual kinase activity and the mammalian target of rapamycin complex 2 (mTORC2) commands this process by phosphorylating AKT at Ser473. Here we showed that embryonic and postnatal lymphatic deletion of Rictor , a critical component of mTORC2, led to a significant decrease in lymphatic valves and prevented the maturation of collecting lymphatic vessels. RICTOR knockdown in human lymphatic endothelial cells (hdLECs) not only significantly reduced the level of activated AKT and the expression of valve-forming genes under no-flow conditions, but also abolished the upregulation of AKT activity and valve-forming genes in response to flow. We further showed that the AKT target, FOXO1, a repressor of lymphatic valve formation, had increased nuclear activity in Rictor knockout mesenteric LECs, in vivo . Deletion of Foxo1 in Rictor knockout mice restored the number of valves to control levels in both mesenteric and ear lymphatics. Our work revealed a novel role of RICTOR signaling in the mechanotransduction signaling pathway, wherein it activates AKT and prevents the nuclear accumulation of the valve repressor, FOXO1, which ultimately allows the formation and maintenance of a normal lymphatic valve.
Collapse
|
31
|
Weigel C, Bellaci J, Spiegel S. Sphingosine-1-phosphate and its receptors in vascular endothelial and lymphatic barrier function. J Biol Chem 2023; 299:104775. [PMID: 37142226 PMCID: PMC10220486 DOI: 10.1016/j.jbc.2023.104775] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/06/2023] Open
Abstract
The vascular and lymphatic systems both comprise a series of structurally distinct vessels lined with an inner layer of endothelial cells that function to provide a semipermeable barrier to blood and lymph. Regulation of the endothelial barrier is critical for maintaining vascular and lymphatic barrier homeostasis. One of the regulators of endothelial barrier function and integrity is sphingosine-1-phosphate (S1P), a bioactive sphingolipid metabolite secreted into the blood by erythrocytes, platelets, and endothelial cells and into the lymph by lymph endothelial cells. Binding of S1P to its G protein-coupled receptors, known as S1PR1-5, regulates its pleiotropic functions. This review outlines the structural and functional differences between vascular and lymphatic endothelium and describes current understanding of the importance of S1P/S1PR signaling in regulation of barrier functions. Most studies thus far have been primarily focused on the role of the S1P/S1PR1 axis in vasculature and have been summarized in several excellent reviews, and thus, we will only discuss new perspectives on the molecular mechanisms of action of S1P and its receptors. Much less is known about the responses of the lymphatic endothelium to S1P and the functions of S1PRs in lymph endothelial cells, and this is the major focus of this review. We also discuss current knowledge related to signaling pathways and factors regulated by the S1P/S1PR axis that control lymphatic endothelial cell junctional integrity. Gaps and limitations in current knowledge are highlighted together with the need to further understand the role of S1P receptors in the lymphatic system.
Collapse
Affiliation(s)
- Cynthia Weigel
- Department of Biochemistry and Molecular Biology Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Jacqueline Bellaci
- Department of Biochemistry and Molecular Biology Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA.
| |
Collapse
|
32
|
Zhao X, Kiyozuka K, Konishi A, Kawabata-Iwakawa R, Minamishima YA, Obinata H. Actin-binding protein Filamin B regulates the cell-surface retention of endothelial sphingosine 1-phosphate receptor 1. J Biol Chem 2023:104851. [PMID: 37220855 PMCID: PMC10300261 DOI: 10.1016/j.jbc.2023.104851] [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: 02/03/2023] [Revised: 05/09/2023] [Accepted: 05/11/2023] [Indexed: 05/25/2023] Open
Abstract
Sphingosine 1-phosphate receptor 1 (S1PR1) is a G protein-coupled receptor essential for vascular development and postnatal vascular homeostasis. When exposed to sphingosine 1-phosphate (S1P) in the blood of ∼1 μM, S1PR1 in endothelial cells retains cell-surface localization, while lymphocyte S1PR1 shows almost complete internalization, suggesting the cell-surface retention of S1PR1 is endothelial cell-specific. To identify regulating factors that function to retain S1PR1 on the endothelial cell surface, here we utilized an enzyme-catalyzed proximity labeling technique followed by proteomic analyses. We identified Filamin B (FLNB), an actin-binding protein involved in F-actin cross-linking, as a candidate regulating protein. We show FLNB knockdown by RNA interference induced massive internalization of S1PR1 into early endosomes, which was partially ligand-dependent and required receptor phosphorylation. Further investigation showed FLNB was also important for the recycling of internalized S1PR1 back to the cell surface. FLNB knockdown did not affect the localization of S1PR3, another S1P receptor subtype expressed in endothelial cells, nor did it affect localization of ectopically expressed β2-adrenergic receptor. Functionally, we show FLNB knockdown in endothelial cells impaired S1P-induced intracellular phosphorylation events and directed cell migration and enhancement of the vascular barrier. Taken together, our results demonstrate that FLNB is a novel regulator critical for S1PR1 cell-surface localization and thereby proper endothelial cell function.
Collapse
Affiliation(s)
- Xian Zhao
- Department of Biochemistry, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Keisuke Kiyozuka
- Department of Biochemistry, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Akimitsu Konishi
- Department of Biochemistry, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Reika Kawabata-Iwakawa
- Division of Integrated Oncology Research, Gunma University Initiative for Advanced Research, Gunma University, Gunma, Japan
| | | | - Hideru Obinata
- Education and Research Support Center, Gunma University Graduate School of Medicine, Gunma, Japan.
| |
Collapse
|
33
|
Kihara Y, Chun J. Molecular and neuroimmune pharmacology of S1P receptor modulators and other disease-modifying therapies for multiple sclerosis. Pharmacol Ther 2023; 246:108432. [PMID: 37149155 DOI: 10.1016/j.pharmthera.2023.108432] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/25/2023] [Accepted: 05/02/2023] [Indexed: 05/08/2023]
Abstract
Multiple sclerosis (MS) is a neurological, immune-mediated demyelinating disease that affects people in the prime of life. Environmental, infectious, and genetic factors have been implicated in its etiology, although a definitive cause has yet to be determined. Nevertheless, multiple disease-modifying therapies (DMTs: including interferons, glatiramer acetate, fumarates, cladribine, teriflunomide, fingolimod, siponimod, ozanimod, ponesimod, and monoclonal antibodies targeting ITGA4, CD20, and CD52) have been developed and approved for the treatment of MS. All the DMTs approved to date target immunomodulation as their mechanism of action (MOA); however, the direct effects of some DMTs on the central nervous system (CNS), particularly sphingosine 1-phosphate (S1P) receptor (S1PR) modulators, implicate a parallel MOA that may also reduce neurodegenerative sequelae. This review summarizes the currently approved DMTs for the treatment of MS and provides details and recent advances in the molecular pharmacology, immunopharmacology, and neuropharmacology of S1PR modulators, with a special focus on the CNS-oriented, astrocyte-centric MOA of fingolimod.
Collapse
Affiliation(s)
- Yasuyuki Kihara
- Sanford Burnham Prebys Medical Discovery Institute, United States of America.
| | - Jerold Chun
- Sanford Burnham Prebys Medical Discovery Institute, United States of America
| |
Collapse
|
34
|
Hashemi E, Yoseph E, Tsai HC, Moreno M, Yeh LH, Mehta SB, Kono M, Proia R, Han MH. Visualizing Sphingosine-1-Phosphate Receptor 1(S1P 1) Signaling During Central Nervous System De- and Remyelination. Cell Mol Neurobiol 2023; 43:1219-1236. [PMID: 35917044 PMCID: PMC10444542 DOI: 10.1007/s10571-022-01245-0] [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: 09/06/2021] [Accepted: 06/14/2022] [Indexed: 11/24/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory-demyelinating disease of the central nervous system (CNS) mediated by aberrant auto-reactive immune responses. The current immune-modulatory therapies are unable to protect and repair immune-mediated neural tissue damage. One of the therapeutic targets in MS is the sphingosine-1-phosphate (S1P) pathway which signals via sphingosine-1-phosphate receptors 1-5 (S1P1-5). S1P receptors are expressed predominantly on immune and CNS cells. Considering the potential neuroprotective properties of S1P signaling, we utilized S1P1-GFP (Green fluorescent protein) reporter mice in the cuprizone-induced demyelination model to investigate in vivo S1P - S1P1 signaling in the CNS. We observed S1P1 signaling in a subset of neural stem cells in the subventricular zone (SVZ) during demyelination. During remyelination, S1P1 signaling is expressed in oligodendrocyte progenitor cells in the SVZ and mature oligodendrocytes in the medial corpus callosum (MCC). In the cuprizone model, we did not observe S1P1 signaling in neurons and astrocytes. We also observed β-arrestin-dependent S1P1 signaling in lymphocytes during demyelination and CNS inflammation. Our findings reveal β-arrestin-dependent S1P1 signaling in oligodendrocyte lineage cells implying a role of S1P1 signaling in remyelination.
Collapse
Affiliation(s)
- Ezzat Hashemi
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, 1201 Welch Rd, MSLS BLG P212, Stanford, CA, 94305, USA
| | - Ezra Yoseph
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, 1201 Welch Rd, MSLS BLG P212, Stanford, CA, 94305, USA
| | - Hsing-Chuan Tsai
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, 1201 Welch Rd, MSLS BLG P212, Stanford, CA, 94305, USA
| | - Monica Moreno
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, 1201 Welch Rd, MSLS BLG P212, Stanford, CA, 94305, USA
| | - Li-Hao Yeh
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | | | - Mari Kono
- Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
| | - Richard Proia
- Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
| | - May H Han
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, 1201 Welch Rd, MSLS BLG P212, Stanford, CA, 94305, USA.
| |
Collapse
|
35
|
van Echten-Deckert G. The role of sphingosine 1-phosphate metabolism in brain health and disease. Pharmacol Ther 2023; 244:108381. [PMID: 36907249 DOI: 10.1016/j.pharmthera.2023.108381] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/02/2023] [Accepted: 03/06/2023] [Indexed: 03/13/2023]
Abstract
Lipids are essential structural and functional components of the central nervous system (CNS). Sphingolipids are ubiquitous membrane components which were discovered in the brain in the late 19th century. In mammals, the brain contains the highest concentration of sphingolipids in the body. Sphingosine 1-phosphate (S1P) derived from membrane sphingolipids evokes multiple cellular responses which, depending on its concentration and localization, make S1P a double-edged sword in the brain. In the present review we highlight the role of S1P in brain development and focus on the often contrasting findings regarding its contributions to the initiation, progression and potential recovery of different brain pathologies, including neurodegeneration, multiple sclerosis (MS), brain cancers, and psychiatric illnesses. A detailed understanding of the critical implications of S1P in brain health and disease may open the door for new therapeutic options. Thus, targeting S1P-metabolizing enzymes and/or signaling pathways might help overcome, or at least ameliorate, several brain illnesses.
Collapse
|
36
|
Zhang L, Liu J, Xiao E, Han Q, Wang L. Sphingosine-1-phosphate related signalling pathways manipulating virus replication. Rev Med Virol 2023; 33:e2415. [PMID: 36597202 DOI: 10.1002/rmv.2415] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 11/28/2022] [Accepted: 11/28/2022] [Indexed: 01/05/2023]
Abstract
Viruses can create a unique cellular environment that facilitates replication and transmission. Sphingosine kinases (SphKs) produce sphingosine-1-phosphate (S1P), a bioactive sphingolipid molecule that performs both physiological and pathological effects primarily by activating a subgroup of the endothelial differentiation gene family of G-protein coupled cell surface receptors known as S1P receptors (S1PR1-5). A growing body of evidence indicates that the SphK/S1P axis is crucial for regulating cellular activities in virus infections like respiratory viruses, enteroviruses, hepatitis viruses, herpes viruses, and arboviruses replicate. Depending on the type of virus, pro- or anti-viral activities of the SphK/S1P axis sometimes rely on the host immune system and sometimes directly through intracellular signalling pathways or cell proliferation. Recent research has shown novel roles of S1P and SphK in viral replication. Sphingosine kinase isoforms (SphK1 and SphK2) levels can be manipulated by several viruses to promote the effects that are expected. Regulation of cellular signalling pathways plays a significant role in the mechanism. The purpose of this review is to provide insight of the characters played by the SphK/S1P axis throughout diverse viral infection processes. We then assess potential therapeutic methods that are based on S1P signalling and metabolism during viral infections.
Collapse
Affiliation(s)
- Lu Zhang
- Center of Clinical Laboratory, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, China
| | - Juan Liu
- Center of Clinical Laboratory, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, China
| | - Erya Xiao
- Center of Clinical Laboratory, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, China
| | - Qingzhen Han
- Center of Clinical Laboratory, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, China
| | - Lin Wang
- Center of Clinical Laboratory, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, China
| |
Collapse
|
37
|
Alam S, Afsar SY, Van Echten-Deckert G. S1P Released by SGPL1-Deficient Astrocytes Enhances Astrocytic ATP Production via S1PR 2,4, Thus Keeping Autophagy in Check: Potential Consequences for Brain Health. Int J Mol Sci 2023; 24:ijms24054581. [PMID: 36902011 PMCID: PMC10003137 DOI: 10.3390/ijms24054581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 02/22/2023] [Accepted: 02/24/2023] [Indexed: 03/02/2023] Open
Abstract
Astrocytes are critical players in brain health and disease. Sphingosine-1-phosphate (S1P), a bioactive signaling lipid, is involved in several vital processes, including cellular proliferation, survival, and migration. It was shown to be crucial for brain development. Its absence is embryonically lethal, affecting, inter alia, the anterior neural tube closure. However, an excess of S1P due to mutations in S1P-lyase (SGPL1), the enzyme responsible for its constitutive removal, is also harmful. Of note, the gene SGPL1 maps to a region prone to mutations in several human cancers and also in S1P-lyase insufficiency syndrome (SPLIS) characterized by several symptoms, including peripheral and central neurological defects. Here, we investigated the impact of S1P on astrocytes in a mouse model with the neural-targeted ablation of SGPL1. We found that SGPL1 deficiency, and hence the accumulation of its substrate, S1P, causes the elevated expression of glycolytic enzymes and preferentially directs pyruvate into the tricarboxylic acid (TCA) cycle through its receptors (S1PR2,4). In addition, the activity of TCA regulatory enzymes was increased, and consequently, so was the cellular ATP content. The high energy load activates the mammalian target of rapamycin (mTOR), thus keeping astrocytic autophagy in check. Possible consequences for the viability of neurons are discussed.
Collapse
|
38
|
Garnier O, Vilgrain I. Dialogue between VE-Cadherin and Sphingosine 1 Phosphate Receptor1 (S1PR1) for Protecting Endothelial Functions. Int J Mol Sci 2023; 24:ijms24044018. [PMID: 36835432 PMCID: PMC9959973 DOI: 10.3390/ijms24044018] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/09/2023] [Accepted: 02/13/2023] [Indexed: 02/19/2023] Open
Abstract
The endothelial cells (EC) of established blood vessels in adults remain extraordinarily quiescent in the sense that they are not actively proliferating, but they fulfill the necessary role to control the permeability of their monolayer that lines the interior of blood vessels. The cell-cell junctions between ECs in the endothelium comprise tight junctions and adherens homotypic junctions, which are ubiquitous along the vascular tree. Adherens junctions are adhesive intercellular contacts that are crucial for the organization of the EC monolayer and its maintenance and regulation of normal microvascular function. The molecular components and underlying signaling pathways that control the association of adherens junctions have been described in the last few years. In contrast, the role that dysfunction of these adherens junctions has in contributing to human vascular disease remains an important open issue. Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid mediator found at high concentrations in blood which has important roles in the control of the vascular permeability, cell recruitment, and clotting that follow inflammatory processes. This role of S1P is achieved through a signaling pathway mediated through a family of G protein-coupled receptors designated as S1PR1. This review highlights novel evidence for a direct linkage between S1PR1 signaling and the mediation of EC cohesive properties that are controlled by VE-cadherin.
Collapse
|
39
|
An Update of G-Protein-Coupled Receptor Signaling and Its Deregulation in Gastric Carcinogenesis. Cancers (Basel) 2023; 15:cancers15030736. [PMID: 36765694 PMCID: PMC9913146 DOI: 10.3390/cancers15030736] [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: 11/03/2022] [Revised: 01/15/2023] [Accepted: 01/19/2023] [Indexed: 01/27/2023] Open
Abstract
G-protein-coupled receptors (GPCRs) belong to a cell surface receptor superfamily responding to a wide range of external signals. The binding of extracellular ligands to GPCRs activates a heterotrimeric G protein and triggers the production of numerous secondary messengers, which transduce the extracellular signals into cellular responses. GPCR signaling is crucial and imperative for maintaining normal tissue homeostasis. High-throughput sequencing analyses revealed the occurrence of the genetic aberrations of GPCRs and G proteins in multiple malignancies. The altered GPCRs/G proteins serve as valuable biomarkers for early diagnosis, prognostic prediction, and pharmacological targets. Furthermore, the dysregulation of GPCR signaling contributes to tumor initiation and development. In this review, we have summarized the research progress of GPCRs and highlighted their mechanisms in gastric cancer (GC). The aberrant activation of GPCRs promotes GC cell proliferation and metastasis, remodels the tumor microenvironment, and boosts immune escape. Through deep investigation, novel therapeutic strategies for targeting GPCR activation have been developed, and the final aim is to eliminate GPCR-driven gastric carcinogenesis.
Collapse
|
40
|
Martín-Hernández D, Martínez M, Robledo-Montaña J, Muñoz-López M, Virto L, Ambrosio N, Marín MJ, Montero E, Herrera D, Sanz M, Leza JC, Figuero E, García-Bueno B. Neuroinflammation related to the blood-brain barrier and sphingosine-1-phosphate in a pre-clinical model of periodontal diseases and depression in rats. J Clin Periodontol 2023; 50:642-656. [PMID: 36644813 DOI: 10.1111/jcpe.13780] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 01/02/2023] [Accepted: 01/03/2023] [Indexed: 01/17/2023]
Abstract
AIM To explore the potential mechanisms of neuroinflammation (microglia, blood-brain barrier [BBB] permeability, and the sphingosine-1-phosphate [S1P] pathways) resulting from the association between periodontitis and depression in rats. MATERIALS AND METHODS This pre-clinical in vivo experimental study used Wistar rats, in which experimental periodontitis (P) was induced by using oral gavages with Porphyromonas gingivalis and Fusobacterium nucleatum. Then, a chronic mild stress (CMS) model was implemented to induce a depressive-like behaviour, resulting in four groups: P with CMS (P+CMS+), P without CMS (P+CMS-), CMS without P (P-CMS+), and control (P-CMS-). After harvesting brain samples, protein/mRNA expression analyses and fluorescence immunohistochemistry were performed in the frontal cortex (FC). Results were analysed by ANOVA. RESULTS CMS exposure increased the number of microglia (an indicator of neuroinflammation) in the FC. In the combined model (P+CMS+), there was a decrease in the expression of tight junction proteins (zonula occludens-1 [ZO-1], occludin) and an increase in intercellular and vascular cell adhesion molecules (ICAM-1, VCAM-1) and matrix metalloproteinase 9 (MMP9), suggesting a more severe disruption of the BBB. The enzymes and receptors of S1P were also differentially regulated. CONCLUSIONS Microglia, BBB permeability, and S1P pathways could be relevant mechanisms explaining the association between periodontitis and depression.
Collapse
Affiliation(s)
- David Martín-Hernández
- Department of Pharmacology and Toxicology, Faculty of Medicine, Complutense University of Madrid (UCM), Hospital 12 de Octubre Research Institute (Imas12), Neurochemistry Research Institute UCM (IUIN), Madrid, Spain.,Biomedical Network Research Center of Mental Health (CIBERSAM), Institute of Health Carlos III, Madrid, Spain
| | - María Martínez
- ETEP (Etiology and Therapy of Periodontal and Peri-implant Diseases) Research Group, UCM, Madrid, Spain.,Department of Dental Clinical Specialties, Faculty of Dentistry, UCM, Madrid, Spain
| | - Javier Robledo-Montaña
- Department of Pharmacology and Toxicology, Faculty of Medicine, Complutense University of Madrid (UCM), Hospital 12 de Octubre Research Institute (Imas12), Neurochemistry Research Institute UCM (IUIN), Madrid, Spain.,Biomedical Network Research Center of Mental Health (CIBERSAM), Institute of Health Carlos III, Madrid, Spain
| | - Marina Muñoz-López
- Department of Pharmacology and Toxicology, Faculty of Medicine, Complutense University of Madrid (UCM), Hospital 12 de Octubre Research Institute (Imas12), Neurochemistry Research Institute UCM (IUIN), Madrid, Spain.,Biomedical Network Research Center of Mental Health (CIBERSAM), Institute of Health Carlos III, Madrid, Spain
| | - Leire Virto
- ETEP (Etiology and Therapy of Periodontal and Peri-implant Diseases) Research Group, UCM, Madrid, Spain.,Department of Anatomy and Embryology, Faculty of Optics, UCM, Madrid, Spain
| | - Nagore Ambrosio
- ETEP (Etiology and Therapy of Periodontal and Peri-implant Diseases) Research Group, UCM, Madrid, Spain.,Department of Dental Clinical Specialties, Faculty of Dentistry, UCM, Madrid, Spain
| | - Maria José Marín
- ETEP (Etiology and Therapy of Periodontal and Peri-implant Diseases) Research Group, UCM, Madrid, Spain
| | - Eduardo Montero
- ETEP (Etiology and Therapy of Periodontal and Peri-implant Diseases) Research Group, UCM, Madrid, Spain.,Department of Dental Clinical Specialties, Faculty of Dentistry, UCM, Madrid, Spain
| | - David Herrera
- ETEP (Etiology and Therapy of Periodontal and Peri-implant Diseases) Research Group, UCM, Madrid, Spain.,Department of Dental Clinical Specialties, Faculty of Dentistry, UCM, Madrid, Spain
| | - Mariano Sanz
- ETEP (Etiology and Therapy of Periodontal and Peri-implant Diseases) Research Group, UCM, Madrid, Spain.,Department of Dental Clinical Specialties, Faculty of Dentistry, UCM, Madrid, Spain
| | - Juan C Leza
- Department of Pharmacology and Toxicology, Faculty of Medicine, Complutense University of Madrid (UCM), Hospital 12 de Octubre Research Institute (Imas12), Neurochemistry Research Institute UCM (IUIN), Madrid, Spain.,Biomedical Network Research Center of Mental Health (CIBERSAM), Institute of Health Carlos III, Madrid, Spain
| | - Elena Figuero
- ETEP (Etiology and Therapy of Periodontal and Peri-implant Diseases) Research Group, UCM, Madrid, Spain.,Department of Dental Clinical Specialties, Faculty of Dentistry, UCM, Madrid, Spain
| | - Borja García-Bueno
- Department of Pharmacology and Toxicology, Faculty of Medicine, Complutense University of Madrid (UCM), Hospital 12 de Octubre Research Institute (Imas12), Neurochemistry Research Institute UCM (IUIN), Madrid, Spain.,Biomedical Network Research Center of Mental Health (CIBERSAM), Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
41
|
Sasset L, Chowdhury KH, Manzo OL, Rubinelli L, Konrad C, Maschek JA, Manfredi G, Holland WL, Di Lorenzo A. Sphingosine-1-phosphate controls endothelial sphingolipid homeostasis via ORMDL. EMBO Rep 2023; 24:e54689. [PMID: 36408842 PMCID: PMC9827560 DOI: 10.15252/embr.202254689] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 10/04/2022] [Accepted: 10/28/2022] [Indexed: 11/22/2022] Open
Abstract
Disruption of sphingolipid homeostasis and signaling has been implicated in diabetes, cancer, cardiometabolic, and neurodegenerative disorders. Yet, mechanisms governing cellular sensing and regulation of sphingolipid homeostasis remain largely unknown. In yeast, serine palmitoyltransferase, catalyzing the first and rate-limiting step of sphingolipid de novo biosynthesis, is negatively regulated by Orm1 and 2. Lowering sphingolipids triggers Orms phosphorylation, upregulation of serine palmitoyltransferase activity and sphingolipid de novo biosynthesis. However, mammalian orthologs ORMDLs lack the N-terminus hosting the phosphosites. Thus, which sphingolipid(s) are sensed by the cells, and mechanisms of homeostasis remain largely unknown. Here, we identify sphingosine-1-phosphate (S1P) as key sphingolipid sensed by cells via S1PRs to maintain homeostasis. The increase in S1P-S1PR signaling stabilizes ORMDLs, restraining SPT activity. Mechanistically, the hydroxylation of ORMDLs at Pro137 allows a constitutive degradation of ORMDLs via ubiquitin-proteasome pathway, preserving SPT activity. Disrupting S1PR/ORMDL axis results in ceramide accrual, mitochondrial dysfunction, impaired signal transduction, all underlying endothelial dysfunction, early event in the onset of cardio- and cerebrovascular diseases. Our discovery may provide the molecular basis for therapeutic intervention restoring sphingolipid homeostasis.
Collapse
Affiliation(s)
- Linda Sasset
- Department of Pathology and Laboratory MedicineCardiovascular Research Institute, Weill Cornell MedicineNew YorkNYUSA
- Brain and Mind Research Institute, Weill Cornell MedicineNew YorkNYUSA
| | - Kamrul H Chowdhury
- Department of Nutrition and Integrative PhysiologyUniversity of Utah College of HealthSalt Lake CityUTUSA
| | - Onorina L Manzo
- Department of Pathology and Laboratory MedicineCardiovascular Research Institute, Weill Cornell MedicineNew YorkNYUSA
- Brain and Mind Research Institute, Weill Cornell MedicineNew YorkNYUSA
- Department of PharmacyUniversity of Naples “Federico II”NaplesItaly
| | - Luisa Rubinelli
- Department of Pathology and Laboratory MedicineCardiovascular Research Institute, Weill Cornell MedicineNew YorkNYUSA
- Brain and Mind Research Institute, Weill Cornell MedicineNew YorkNYUSA
| | - Csaba Konrad
- Department of Nutrition and Integrative PhysiologyUniversity of Utah College of HealthSalt Lake CityUTUSA
| | - J Alan Maschek
- Department of Nutrition and Integrative PhysiologyUniversity of Utah College of HealthSalt Lake CityUTUSA
| | - Giovanni Manfredi
- Brain and Mind Research Institute, Weill Cornell MedicineNew YorkNYUSA
| | - William L Holland
- Department of Nutrition and Integrative PhysiologyUniversity of Utah College of HealthSalt Lake CityUTUSA
| | - Annarita Di Lorenzo
- Department of Pathology and Laboratory MedicineCardiovascular Research Institute, Weill Cornell MedicineNew YorkNYUSA
- Brain and Mind Research Institute, Weill Cornell MedicineNew YorkNYUSA
| |
Collapse
|
42
|
Levesque MV, Hla T. Signal Transduction and Gene Regulation in the Endothelium. Cold Spring Harb Perspect Med 2023; 13:a041153. [PMID: 35667710 PMCID: PMC9722983 DOI: 10.1101/cshperspect.a041153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Extracellular signals act on G-protein-coupled receptors (GPCRs) to regulate homeostasis and adapt to stress. This involves rapid intracellular post-translational responses and long-lasting gene-expression changes that ultimately determine cellular phenotype and fate changes. The lipid mediator sphingosine 1-phosphate (S1P) and its receptors (S1PRs) are examples of well-studied GPCR signaling axis essential for vascular development, homeostasis, and diseases. The biochemical cascades involved in rapid S1P signaling are well understood. However, gene-expression regulation by S1PRs are less understood. In this review, we focus our attention to how S1PRs regulate nuclear chromatin changes and gene transcription to modulate vascular and lymphatic endothelial phenotypic changes during embryonic development and adult homeostasis. Because S1PR-targeted drugs approved for use in the treatment of autoimmune diseases cause substantial vascular-related adverse events, these findings are critical not only for general understanding of stimulus-evoked gene regulation in the vascular endothelium, but also for therapeutic development of drugs for autoimmune and perhaps vascular diseases.
Collapse
Affiliation(s)
- Michel V Levesque
- Vascular Biology Program, Boston Children's Hospital and Department of Surgery, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Timothy Hla
- Vascular Biology Program, Boston Children's Hospital and Department of Surgery, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
43
|
Yadunandanan Nair N, Samuel V, Ramesh L, Marib A, David DT, Sundararaman A. Actin cytoskeleton in angiogenesis. Biol Open 2022; 11:bio058899. [PMID: 36444960 PMCID: PMC9729668 DOI: 10.1242/bio.058899] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2024] Open
Abstract
Actin, one of the most abundant intracellular proteins in mammalian cells, is a critical regulator of cell shape and polarity, migration, cell division, and transcriptional response. Angiogenesis, or the formation of new blood vessels in the body is a well-coordinated multi-step process. Endothelial cells lining the blood vessels acquire several new properties such as front-rear polarity, invasiveness, rapid proliferation and motility during angiogenesis. This is achieved by changes in the regulation of the actin cytoskeleton. Actin remodelling underlies the switch between the quiescent and angiogenic state of the endothelium. Actin forms endothelium-specific structures that support uniquely endothelial functions. Actin regulators at endothelial cell-cell junctions maintain the integrity of the blood-tissue barrier while permitting trans-endothelial leukocyte migration. This review focuses on endothelial actin structures and less-recognised actin-mediated endothelial functions. Readers are referred to other recent reviews for the well-recognised roles of actin in endothelial motility, barrier functions and leukocyte transmigration. Actin generates forces that are transmitted to the extracellular matrix resulting in vascular matrix remodelling. In this review, we attempt to synthesize our current understanding of the roles of actin in vascular morphogenesis. We speculate on the vascular bed specific differences in endothelial actin regulation and its role in the vast heterogeneity in endothelial morphology and function across the various tissues of our body.
Collapse
Affiliation(s)
- Nidhi Yadunandanan Nair
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India695014
| | - Victor Samuel
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India695014
| | - Lariza Ramesh
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India695014
| | - Areeba Marib
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India695014
| | - Deena T. David
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India695014
| | - Ananthalakshmy Sundararaman
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India695014
| |
Collapse
|
44
|
Lee HJ, Choe K, Park JS, Khan A, Kim MW, Park TJ, Kim MO. O-Cyclic Phytosphingosine-1-Phosphate Protects against Motor Dysfunctions and Glial Cell Mediated Neuroinflammation in the Parkinson's Disease Mouse Models. Antioxidants (Basel) 2022; 11:2107. [PMID: 36358479 PMCID: PMC9686509 DOI: 10.3390/antiox11112107] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/14/2022] [Accepted: 10/22/2022] [Indexed: 10/29/2023] Open
Abstract
O-cyclic phytosphingosine-1-phosphate (cPS1P) is a novel and chemically synthesized sphingosine metabolite derived from phytosphingosine-1-phosphate (S1P). This study was undertaken to unveil the potential neuroprotective effects of cPS1P on two different mouse models of Parkinson's disease (PD). The study used 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and neuron specific enolase promoter human alpha-synuclein (NSE-hαSyn) Korl transgenic mice. MPTP was injected for five consecutive days and cPS1P was injected for alternate days for six weeks intraperitoneally. We performed behavioral tests and analyzed the immunohistochemistry and immunofluorescence staining in the substantia nigra pars compacta (SNpc) and the striatum. The behavior tests showed a significant reduction in the motor functions in the PD models, which was reversed with the administration of cPS1P. In addition, both PD-models showed reduced expression of the sphingosine-1-phosphate receptor 1 (S1PR1), and α-Syn which was restored with cPS1P treatment. In addition, administration of cPS1P restored dopamine-related proteins such as tyrosine hydroxylase (TH), vesicular monoamine transporter 2 (VMAT2), and dopamine transporter (DAT). Lastly, neuroinflammatory related markers such as glial fibrillary acidic protein (GFAP), ionized calcium-binding adapter protein-1 (Iba-1), c-Jun N-terminal kinases (JNK), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB), tumor necrosis factor-alpha (TNF-α), and interleukin 1 beta (IL-1β) were all reduced after cPS1P administration. The overall findings supported the notion that cPS1P protects against dopamine depletion, neuroinflammation, and PD-associated symptoms.
Collapse
Affiliation(s)
- Hyeon Jin Lee
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea
| | - Kyonghwan Choe
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, 6229ER Maastricht, The Netherlands
| | - Jun Sung Park
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea
| | - Amjad Khan
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea
| | - Min Woo Kim
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea
| | - Tae Ju Park
- Haemato-oncology/Systems Medicine Group, Paul O’Gorman Leukaemia Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary & Life Sciences (MVLS), University of Glasgow, Glasgow G12 0ZD, UK
| | - Myeong Ok Kim
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea
- Alz-Dementia Korea Co., Jinju 52828, Korea
| |
Collapse
|
45
|
Hasan Z, Nguyen TQ, Lam BWS, Wong JHX, Wong CCY, Tan CKH, Yu J, Thiam CH, Zhang Y, Angeli V, Nguyen LN. Postnatal deletion of Spns2 prevents neuroinflammation without compromising blood vascular functions. Cell Mol Life Sci 2022; 79:541. [PMID: 36198832 PMCID: PMC11802987 DOI: 10.1007/s00018-022-04573-y] [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: 01/04/2022] [Revised: 09/09/2022] [Accepted: 09/27/2022] [Indexed: 11/03/2022]
Abstract
Protein Spinster homolog 2 (Spns2) is a sphingosine-1-phosphate (S1P) transporter that releases S1P to regulate lymphocyte egress and trafficking. Global deletion of Spns2 (Spns2-/-) has been shown to reduce disease severity in several autoimmune disease models. To examine whether Spns2 could be exploited as a drug target, we generated and characterized the mice with postnatal knockout of Spns2 (Spns2-Mx1Cre). Our results showed that Spns2-Mx1Cre mice had significantly low number of lymphocytes in blood and lymphoid organs similar to Spns2-/- mice. Lymph but not plasma S1P levels were significantly reduced in both groups of knockout mice. Our lipidomic results also showed that Spns2 releases different S1P species into lymph. Interestingly, lymphatic vessels in the lymph nodes (LNs) of Spns2-/- and Spns2-Mx1Cre mice exhibited morphological defects. The structures of high endothelial venules (HEV) in the LNs of Spns2-Mx1Cre mice were disorganized. These results indicate that lack of Spns2 affects both S1P secretion and LN vasculatures. Nevertheless, blood vasculature of these Spns2 deficient mice was not different to controls under homeostasis and vascular insults. Importantly, Spns2-Mx1Cre mice were resistant to multiple sclerosis in experimental autoimmune encephalomyelitis (EAE) models with significant reduction of pathogenic Th17 cells in the central nervous system (CNS). This study suggests that pharmacological inhibition of Spns2 may be exploited for therapeutic applications in treatment of neuroinflammation.
Collapse
Affiliation(s)
- Zafrul Hasan
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Toan Q Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Brenda Wan Shing Lam
- Department of Pharmacology, Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore, Singapore
| | - Jovi Hui Xin Wong
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Caleb Cheng Yi Wong
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Clarissa Kai Hui Tan
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Jiabo Yu
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- Life Sciences Institute, Immunology Program, National University of Singapore, Singapore, 117456, Singapore
- Immunology Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
| | - Chung Hwee Thiam
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- Life Sciences Institute, Immunology Program, National University of Singapore, Singapore, 117456, Singapore
- Immunology Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
| | - Yongliang Zhang
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- Life Sciences Institute, Immunology Program, National University of Singapore, Singapore, 117456, Singapore
- Immunology Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
| | - Veronique Angeli
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- Life Sciences Institute, Immunology Program, National University of Singapore, Singapore, 117456, Singapore
- Immunology Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
| | - Long N Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore.
- Life Sciences Institute, Singapore Lipidomics Incubator (SLING), National University of Singapore, Singapore, 117456, Singapore.
- Cardiovascular Disease Research (CVD) Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117545, Singapore.
- Life Sciences Institute, Immunology Program, National University of Singapore, Singapore, 117456, Singapore.
- Immunology Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore.
| |
Collapse
|
46
|
Siponimod ameliorates metabolic oligodendrocyte injury via the sphingosine-1 phosphate receptor 5. Proc Natl Acad Sci U S A 2022; 119:e2204509119. [PMID: 36161894 DOI: 10.1073/pnas.2204509119] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Multiple sclerosis (MS), an autoimmune-driven, inflammatory demyelinating disease of the central nervous system (CNS), causes irreversible accumulation of neurological deficits to a variable extent. Although there are potent disease-modifying agents for its initial relapsing-remitting phase, immunosuppressive therapies show limited efficacy in secondary progressive MS (SPMS). Although modulation of sphingosine-1 phosphate receptors has proven beneficial during SPMS, the underlying mechanisms are poorly understood. In this project, we followed the hypothesis that siponimod, a sphingosine-1 phosphate receptor modulator, exerts protective effects by direct modulation of glia cell function (i.e., either astrocytes, microglia, or oligodendrocytes). To this end, we used the toxin-mediated, nonautoimmune MS animal model of cuprizone (Cup) intoxication. On the histological level, siponimod ameliorated cuprizone-induced oligodendrocyte degeneration, demyelination, and axonal injury. Protective effects were evident as well using GE180 translocator protein 18-kDa (TSPO) imaging with positron emission tomography (PET)/computed tomography (CT) imaging or next generation sequencing (NGS). Siponimod also ameliorated the cuprizone-induced pathologies in Rag1-deficient mice, demonstrating that the protection is independent of T and B cell modulation. Proinflammatory responses in primary mixed astrocytes/microglia cell cultures were not modulated by siponimod, suggesting that other cell types than microglia and astrocytes are targeted. Of note, siponimod completely lost its protective effects in S1pr5-deficient mice, suggesting direct protection of degenerating oligodendrocytes. Our study demonstrates that siponimod exerts protective effects in the brain in a S1PR5-dependent manner. This finding is not just relevant in the context of MS but in other neuropathologies as well, characterized by a degeneration of the axon-myelin unit.
Collapse
|
47
|
Paranjpe V, Galor A, Grambergs R, Mandal N. The role of sphingolipids in meibomian gland dysfunction and ocular surface inflammation. Ocul Surf 2022; 26:100-110. [PMID: 35973562 PMCID: PMC10259413 DOI: 10.1016/j.jtos.2022.07.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 07/17/2022] [Accepted: 07/22/2022] [Indexed: 11/26/2022]
Abstract
Inflammation occurs in response to tissue injury and invasion of microorganisms and is carried out by the innate and adaptive immune systems, which are regulated by numerous chemokines, cytokines, and lipid mediators. There are four major families of bioactive lipid mediators that play an integral role in inflammation - eicosanoids, sphingolipids (SPL), specialized pro-resolving mediators (SPM), and endocannabinoids. SPL have been historically recognized as important structural components of cellular membranes; their roles as bioactive lipids and inflammatory mediators are recent additions. Major SPL metabolites, including sphingomyelin, ceramide, ceramide 1-phosphate (C1P), sphingosine, sphingosine 1-phosphate (S1P), and their respective enzymes have been studied extensively, primarily in cell-culture and animal models, for their roles in cellular signaling and regulating inflammation and apoptosis. Less focus has been given to the involvement of SPL in eye diseases. As such, the aim of this review was to examine relationships between the SPL family and ocular surface diseases, focusing on their role in disease pathophysiology and discussing the potential of therapeutics that disrupt SPL pathways.
Collapse
Affiliation(s)
- Vikram Paranjpe
- Department of Ophthalmology, New York University School of Medicine, 550 First Avenue, New York, NY, 10016, USA
| | - Anat Galor
- Miami Veterans Administration Medical Center, 1201 NW 16th St, Miami, FL, 33125, USA; Bascom Palmer Eye Institute, University of Miami, 900 NW 17th Street, Miami, FL, 33136, USA.
| | - Richard Grambergs
- Departments of Ophthalmology, Anatomy and Neurobiology, University of Tennessee Health Sciences Center, Hamilton Eye Institute, 930 Madison Avenue, Memphis, TN, 38163, USA
| | - Nawajes Mandal
- Departments of Ophthalmology, Anatomy and Neurobiology, University of Tennessee Health Sciences Center, Hamilton Eye Institute, 930 Madison Avenue, Memphis, TN, 38163, USA; Memphis VA Medical Center, Memphis, TN, 38104, USA.
| |
Collapse
|
48
|
S1PR1 serves as a viable drug target against pulmonary fibrosis by increasing the integrity of the endothelial barrier of the lung. Acta Pharm Sin B 2022; 13:1110-1127. [PMID: 36970190 PMCID: PMC10031262 DOI: 10.1016/j.apsb.2022.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 09/26/2022] [Accepted: 10/07/2022] [Indexed: 11/20/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive lung disease with unclear etiology and limited treatment options. The median survival time for IPF patients is approximately 2-3 years and there is no effective intervention to treat IPF other than lung transplantation. As important components of lung tissue, endothelial cells (ECs) are associated with pulmonary diseases. However, the role of endothelial dysfunction in pulmonary fibrosis (PF) is incompletely understood. Sphingosine-1-phosphate receptor 1 (S1PR1) is a G protein-coupled receptor highly expressed in lung ECs. Its expression is markedly reduced in patients with IPF. Herein, we generated an endothelial-conditional S1pr1 knockout mouse model which exhibited inflammation and fibrosis with or without bleomycin (BLM) challenge. Selective activation of S1PR1 with an S1PR1 agonist, IMMH002, exerted a potent therapeutic effect in mice with bleomycin-induced fibrosis by protecting the integrity of the endothelial barrier. These results suggest that S1PR1 might be a promising drug target for IPF therapy.
Collapse
|
49
|
Yang C, Liu Y, Wang Z, Lin M, Liu C. Controlled mechanical loading improves bone regeneration by regulating type H vessels in a S1Pr1-dependent manner. FASEB J 2022; 36:e22530. [PMID: 36063128 DOI: 10.1096/fj.202200339rrr] [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: 03/01/2022] [Revised: 08/04/2022] [Accepted: 08/22/2022] [Indexed: 11/11/2022]
Abstract
Despite the best treatment, approximately 10% of fractures still face undesirable repair and result in delayed unions or non-unions. Dynamic mechanical stimulation promotes bone formation, when applied at the correct time frame, with optimal loading magnitude, frequency, and repetition. Controlled mechanical loading significantly increases osteogenic cells during the matrix deposition phase of bone repair. In the bone defect, the blood vessel network guides the initial bone formation activities. A unique blood vessel subtype (Type H) exists in bone, which expresses high levels of CD31 and endomucin, and functions to couple angiogenesis and osteogenesis. However, how this form of controlled mechanical loading regulates the Type H vessels and promotes bone formation is still not clear. Sphingosine 1-phosphate (S1P) participates in the bone anabolic process and is a key regulator of the blood vessel. Its receptor, sphingosine 1-phosphate receptor 1 (S1Pr1), is a mechanosensitive protein that regulates vascular integrity. Therefore, we hypothesis that controlled anabolic mechanical loading promotes bone repair by acting on Type H vessels. To study the effect of S1Pr1 on loading induced-bone repair, we utilized a stabilized tibial defect model, which allows for the application of anabolic mechanical loading. Mechanical loading upregulated S1Pr1 within the entire defect, with up to 80% expressed in blood vessels, as observed by deep tissue imaging. Additionally, S1Pr1 antagonism by W146 inhibited the anabolic effects of mechanical loading. We showed that mechanical loading or activating S1Pr1 could induce YAP nuclear translocation, a key regulator in the cell's mechanical response, in endothelial cells (ECs) in vitro. Inhibition of S1Pr1 in endothelial cells by siRNA reduced loading-induced YAP nuclear translocation and expressions of angiogenic genes. In vivo, YAP nuclear translocation in Type H vessels was up-regulated after mechanical loading but was inhibited by antagonizing S1Pr1. S1Pr1 agonist, FTY720, increased bone volume and Type H vessel volume, similar to that of mechanical stimulation. In conclusion, controlled anabolic mechanical loading enhanced bone formation mainly through Type H vessels in a S1Pr1-dependent manner.
Collapse
Affiliation(s)
- Chengyu Yang
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China.,Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
| | - Yang Liu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China.,Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
| | - Ziyan Wang
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China.,Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
| | - Minmin Lin
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China.,Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
| | - Chao Liu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China.,Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
50
|
Wilkerson JL, Basu SK, Stiles MA, Prislovsky A, Grambergs RC, Nicholas SE, Karamichos D, Allegood JC, Proia RL, Mandal N. Ablation of Sphingosine Kinase 1 Protects Cornea from Neovascularization in a Mouse Corneal Injury Model. Cells 2022; 11:2914. [PMID: 36139489 PMCID: PMC9497123 DOI: 10.3390/cells11182914] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 09/03/2022] [Accepted: 09/10/2022] [Indexed: 11/24/2022] Open
Abstract
The purpose of this study was to investigate the role of sphingosine kinase 1 (SphK1), which generates sphingosine-1-phosphate (S1P), in corneal neovascularization (NV). Wild-type (WT) and Sphk1 knockout (Sphk1-/-) mice received corneal alkali-burn treatment to induce corneal NV by placing a 2 mm round piece of Whatman No. 1 filter paper soaked in 1N NaOH on the center of the cornea for 20 s. Corneal sphingolipid species were extracted and identified using liquid chromatography/mass spectrometry (LC/MS). The total number of tip cells and those positive for ethynyl deoxy uridine (EdU) were quantified. Immunocytochemistry was done to examine whether pericytes were present on newly forming blood vessels. Cytokine signaling and angiogenic markers were compared between the two groups using multiplex assays. Data were analyzed using appropriate statistical tests. Here, we show that ablation of SphK1 can significantly reduce NV invasion in the cornea following injury. Corneal sphingolipid analysis showed that total levels of ceramides, monohexosyl ceramides (HexCer), and sphingomyelin were significantly elevated in Sphk-/- corneas compared to WT corneas, with a comparable level of sphingosine among the two genotypes. The numbers of total and proliferating endothelial tip cells were also lower in the Sphk1-/- corneas following injury. This study underscores the role of S1P in post-injury corneal NV and raises further questions about the roles played by ceramide, HexCer, and sphingomyelin in regulating corneal NV. Further studies are needed to unravel the role played by bioactive sphingolipids in maintenance of corneal transparency and clear vision.
Collapse
Affiliation(s)
- Joseph L. Wilkerson
- Dean A. McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT 84112, USA
| | - Sandip K. Basu
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Sciences Center, Memphis, TN 38163, USA
| | - Megan A. Stiles
- Dean A. McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Amanda Prislovsky
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Sciences Center, Memphis, TN 38163, USA
| | - Richard C. Grambergs
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Sciences Center, Memphis, TN 38163, USA
| | - Sarah E. Nicholas
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
- Department of Pharmaceutical Sciences, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Dimitrios Karamichos
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
- Department of Pharmaceutical Sciences, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Jeremy C. Allegood
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
| | - Richard L. Proia
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nawajes Mandal
- Dean A. McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Sciences Center, Memphis, TN 38163, USA
- Departments of Anatomy and Neurobiology, and Pharmaceutical Sciences, University of Tennessee Health Sciences Center, Memphis, TN 38163, USA
- Memphis VA Medical Center, Memphis, TN 38104, USA
| |
Collapse
|