1
|
Edvinsson C, Piani F, Matthes F, Vanherle L, Erlandsson L, Meissner A, Hansson SR. Sphingosine-1-Phosphate, a Marker of Endothelial Injury and Disease Severity in Preeclampsia. Hypertension 2025; 82:914-925. [PMID: 39840434 PMCID: PMC12002043 DOI: 10.1161/hypertensionaha.124.24118] [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: 10/09/2024] [Accepted: 01/06/2025] [Indexed: 01/23/2025]
Abstract
BACKGROUND Preeclampsia is a hypertensive pregnancy disorder marked by endothelial damage. Healthy endothelium is covered by a protective glycocalyx layer, which, when degraded, releases detectable products into the blood. Sphingosine-1-phosphate (S1P) is a cardiovascular biomarker involved in glycocalyx preservation, linked to placentation and preeclampsia development. The study aimed to test the hypothesis that plasma S1P is altered alongside glycocalyx degradation products in severe preeclampsia compared with controls. METHODS We included 121 females: 41 with severe preeclampsia requiring treatment in the intensive care unit, 40 with preeclampsia but no need of intensive care unit treatment, and 40 with normotensive pregnancies. Plasma levels of S1P and glycocalyx degradation products-hyaluronic acid, SDC-1 (syndecan-1), and HSPG2 (heparan sulfate proteoglycan-2)-were analyzed from blood samples taken within 27 hours postpartum. RESULTS Postpartum plasma S1P was significantly lower in the intensive care unit cohort compared with both preeclampsia controls and normotensive controls (P<0.001). Hyaluronic acid and SDC-1 levels were elevated in the intensive care unit group versus normotensive controls (P=0.009 and P=0.023), while HSPG2 was lower (P<0.001). Plasma S1P correlated with hyaluronic acid and blood pressure. CONCLUSION Intensive care patients with severe preeclampsia have lower plasma S1P levels and higher concentrations of glycocalyx degradation products, indicating more pronounced endothelial damage. These findings suggest that S1P is associated with preeclampsia severity and may serve as a biomarker to assess vascular damage in this patient population. Further studies are needed to explore the potential role of S1P in long-term cardiovascular risk assessment for patients with preeclampsia.
Collapse
Affiliation(s)
- Camilla Edvinsson
- Division of Obstetrics and Gynecology, Institute of Clinical Sciences Lund (C.E., F.P., L.E., S.R.H.), Lund University, Sweden
- Division of Anesthesia and Intensive Care, Institute of Clinical Sciences Lund (C.E.), Lund University, Sweden
- Department of Anesthesia and Intensive Care, Helsingborg Hospital, Sweden (C.E.)
| | - Federica Piani
- Division of Obstetrics and Gynecology, Institute of Clinical Sciences Lund (C.E., F.P., L.E., S.R.H.), Lund University, Sweden
- Department of Medical and Surgical Sciences, University of Bologna, Italy (F.P.)
| | - Frank Matthes
- Department of Experimental Medical Science (F.M., L.V., A.M.), Lund University, Sweden
- Wallenberg Center for Molecular Medicine (F.M., L.V., A.M.), Lund University, Sweden
- Physiology, Institute of Theoretical Medicine, University of Augsburg, Germany (F.M., A.M.)
| | - Lotte Vanherle
- Department of Experimental Medical Science (F.M., L.V., A.M.), Lund University, Sweden
- Wallenberg Center for Molecular Medicine (F.M., L.V., A.M.), Lund University, Sweden
| | - Lena Erlandsson
- Division of Obstetrics and Gynecology, Institute of Clinical Sciences Lund (C.E., F.P., L.E., S.R.H.), Lund University, Sweden
| | - Anja Meissner
- Department of Experimental Medical Science (F.M., L.V., A.M.), Lund University, Sweden
- Wallenberg Center for Molecular Medicine (F.M., L.V., A.M.), Lund University, Sweden
- Physiology, Institute of Theoretical Medicine, University of Augsburg, Germany (F.M., A.M.)
| | - Stefan R. Hansson
- Division of Obstetrics and Gynecology, Institute of Clinical Sciences Lund (C.E., F.P., L.E., S.R.H.), Lund University, Sweden
- Department of Obstetrics and Gynecology, Skåne University Hospital, Lund/Malmö, Sweden (S.R.H.)
| |
Collapse
|
2
|
Jung B, Yagi H, Kuo A, Dorweiler TF, Aikawa M, Kasai T, Singh SA, Dannenberg AJ, Fu Z, Niaudet C, Smith LEH, Hla T. ApoM-bound S1P acts via endothelial S1PR1 to suppress choroidal neovascularization and vascular leakage. Angiogenesis 2025; 28:24. [PMID: 40266369 PMCID: PMC12018641 DOI: 10.1007/s10456-025-09975-7] [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: 12/12/2024] [Accepted: 03/13/2025] [Indexed: 04/24/2025]
Abstract
Neovascular age-related macular degeneration (nAMD) is a major cause of vision loss worldwide. Current standard of care is repetitive intraocular injections of vascular endothelial growth factor (VEGF) inhibitors, although responses may be partial and non-durable. We report that circulating sphingosine 1-phosphate (S1P) carried by apolipoprotein M (ApoM) acts through the endothelial S1P receptor 1 (S1PR1) to suppress choroidal neovascularization (CNV) in mouse laser-induced CNV, modeling nAMD. In humans, low plasma ApoM levels were associated with increased choroidal and retinal pathology. Additionally, endothelial S1pr1 knockout and overexpressing transgenic mice showed increased and reduced CNV lesion size, respectively. Systemic administration of ApoM-Fc, an engineered S1P chaperone protein, not only attenuated CNV to an equivalent degree as anti-VEGF antibody treatment but also suppressed pathological vascular leakage. We suggest that modulating circulating ApoM-bound S1P action on endothelial S1PR1 provides a novel therapeutic strategy to treat nAMD.
Collapse
Affiliation(s)
- Bongnam Jung
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Hitomi Yagi
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Andrew Kuo
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Tim F Dorweiler
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Taku Kasai
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Sasha A Singh
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | | | - Zhongjie Fu
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Colin Niaudet
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA.
- CRCI2NA, INSERM, CNRS, Nantes University, 44000, Nantes, France.
| | - Lois E H Smith
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| | - Timothy Hla
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
3
|
Peng H, Du Z, Li J, Wang W, Li Z, Ru S. The sprouting angiogenesis and vascular dysfunction triggered by bisphenol S and tetrabromobisphenol S through disrupting vascular endothelial-cadherin in zebrafish. ENVIRONMENTAL RESEARCH 2025; 278:121632. [PMID: 40246265 DOI: 10.1016/j.envres.2025.121632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/31/2025] [Accepted: 04/15/2025] [Indexed: 04/19/2025]
Abstract
Exogenous chemical toxicants may be important inducers of pathological angiogenesis diseases. However, few studies have investigated the associations between pathological angiogenesis diseases and chemical toxicant exposures, and the specific mechanism by which chemical toxicants induce sprouting angiogenesis is unclear. In this study, zebrafish were exposed to bisphenol S (BPS, 1-100 μg/L) and tetrabromobisphenol S (TBBPS, 0.1 and 10 μg/L) from the embryonic stage to the larval stage to investigate how pollutants interfere with angiogenesis and the function of ectopic sprouting vessels. The results showed that BPS and TBBPS promoted ectopic sprouting angiogenesis in different types of vascular plexuses, including the posterior cardinal vein (PCV) and superficial choroidal vessels (SOVs), at different developmental time points. Proteomic analyses of eGFP-positive endothelial cells (ECs) isolated from Tg(flk1: eGFP) zebrafish revealed that both BPS and TBBPS induced ectopic angiogenesis by acting on vascular endothelial-cadherin (VE-cadherin) and activating downstream proangiogenic signaling. In ectopic sprouting vessels induced by BPS and TBBPS, increased endothelial permeability resulted in white blood cell recruitment. Human oxidized lipids also tended to deposit in these ectopic vessels following BPS and TBBPS exposure. These findings suggest that chemical toxicant-induced ectopic angiogenesis is an important cause of vascular dysfunction and related diseases.
Collapse
Affiliation(s)
- Hongyuan Peng
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Zehui Du
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Jiali Li
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Weiwei Wang
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China.
| | - Ze Li
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Shaoguo Ru
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| |
Collapse
|
4
|
Zheng H, Yu J, Gao L, Wang K, Xu Z, Zeng Z, Zheng K, Tang X, Tian X, Zhao Q, Zhao J, Wan H, Cao Z, Zhang K, Cheng J, Brosius J, Zhang H, Li W, Yan W, Shao Z, Luo F, Deng C. S1PR1-biased activation drives the resolution of endothelial dysfunction-associated inflammatory diseases by maintaining endothelial integrity. Nat Commun 2025; 16:1826. [PMID: 39979282 PMCID: PMC11842847 DOI: 10.1038/s41467-025-57124-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 02/10/2025] [Indexed: 02/22/2025] Open
Abstract
G protein-coupled sphingosine-1-phosphate receptor 1 (S1PR1), a drug target for inflammatory bowel disease (IBD), enables immune cells to egress from lymph nodes, but the treatment increases the risk of immunosuppression. The functional signaling pathway triggered by S1PR1 activation in endothelial cells and its therapeutic application remains unclear. Here, we showed that S1PR1 is highly expressed in endothelial cells of IBD patients and positively correlated with endothelial markers. Gi-biased agonist-SAR247799 activated S1PR1 and reversed pathology in male mouse and organoid IBD models by protecting the integrity of the endothelial barrier without affecting immune cell egress. Cryo-electron microscopy structure of S1PR1-Gi signaling complex bound to SAR247799 with a resolution of 3.47 Å revealed the recognition mode for the biased ligand. With the efficacy of SAR247799 in treating other endothelial dysfunction-associated inflammatory diseases, our study offers mechanistic insights into the Gi-biased S1PR1 agonist and represents a strategy for endothelial dysfunction-associated disease treatment.
Collapse
Affiliation(s)
- Huaping Zheng
- Department of Respiratory and Critical Care Medicine, Center for High Altitude Medicine, Institutes for Systems Genetics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Jingjing Yu
- Department of Respiratory and Critical Care Medicine, Center for High Altitude Medicine, Institutes for Systems Genetics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- Division of Nephrology and Kidney Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Luhua Gao
- Department of Respiratory and Critical Care Medicine, Center for High Altitude Medicine, Institutes for Systems Genetics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Kexin Wang
- Department of Respiratory and Critical Care Medicine, Center for High Altitude Medicine, Institutes for Systems Genetics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- Division of Nephrology and Kidney Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Zheng Xu
- Department of Respiratory and Critical Care Medicine, Center for High Altitude Medicine, Institutes for Systems Genetics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Zhen Zeng
- Department of Gastroenterology, Lab of Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Kun Zheng
- Department of Respiratory and Critical Care Medicine, Center for High Altitude Medicine, Institutes for Systems Genetics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoju Tang
- Department of Respiratory and Critical Care Medicine, Center for High Altitude Medicine, Institutes for Systems Genetics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaowen Tian
- Department of Respiratory and Critical Care Medicine, Center for High Altitude Medicine, Institutes for Systems Genetics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- Division of Nephrology and Kidney Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Qing Zhao
- Department of Respiratory and Critical Care Medicine, Center for High Altitude Medicine, Institutes for Systems Genetics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Jie Zhao
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, China
| | - Huajing Wan
- Department of Respiratory and Critical Care Medicine, Center for High Altitude Medicine, Institutes for Systems Genetics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Zhongwei Cao
- Department of Respiratory and Critical Care Medicine, Center for High Altitude Medicine, Institutes for Systems Genetics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Kang Zhang
- Center for Biomedicine and Innovations, Faculty of Medicine, Macau University of Science and Technology and University Hospital, Macau, China
| | - Jingqiu Cheng
- Department of Respiratory and Critical Care Medicine, Center for High Altitude Medicine, Institutes for Systems Genetics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Jürgen Brosius
- Department of Respiratory and Critical Care Medicine, Center for High Altitude Medicine, Institutes for Systems Genetics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Hu Zhang
- Department of Gastroenterology, Lab of Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Li
- Department of Dermatology, Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Yan
- Department of Respiratory and Critical Care Medicine, Center for High Altitude Medicine, Institutes for Systems Genetics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, China.
| | - Zhenhua Shao
- Department of Respiratory and Critical Care Medicine, Center for High Altitude Medicine, Institutes for Systems Genetics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.
- Division of Nephrology and Kidney Research Institute, West China Hospital, Sichuan University, Chengdu, China.
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, China.
| | - Fengming Luo
- Department of Respiratory and Critical Care Medicine, Center for High Altitude Medicine, Institutes for Systems Genetics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.
| | - Cheng Deng
- Department of Respiratory and Critical Care Medicine, Center for High Altitude Medicine, Institutes for Systems Genetics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
5
|
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
|
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
|
Del Gaudio I, Nitzsche A, Boyé K, Bonnin P, Poulet M, Nguyen TQ, Couty L, Ha HTT, Nguyen DT, Cazenave-Gassiot A, Ben Alaya K, Thérond P, Chun J, Wenk MR, Proia RL, Henrion D, Nguyen LN, Eichmann A, Camerer E. Zonation and ligand and dose dependence of sphingosine 1-phosphate receptor-1 signalling in blood and lymphatic vasculature. Cardiovasc Res 2024; 120:1794-1810. [PMID: 39086170 PMCID: PMC11587562 DOI: 10.1093/cvr/cvae168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/25/2024] [Accepted: 06/12/2024] [Indexed: 08/02/2024] Open
Abstract
AIMS Circulating levels of sphingosine 1-phosphate (S1P), an HDL-associated ligand for the endothelial cell (EC) protective S1P receptor-1 (S1PR1), are reduced in disease states associated with endothelial dysfunction. Yet, as S1PR1 has high affinity for S1P and can be activated by ligand-independent mechanisms and EC autonomous S1P production, it is unclear if relative reductions in circulating S1P can cause endothelial dysfunction. It is also unclear how EC S1PR1 insufficiency, whether induced by deficiency in circulating ligand or by S1PR1-directed immunosuppressive therapy, affects different vascular subsets. METHODS AND RESULTS We here fine map the zonation of S1PR1 signalling in the murine blood and lymphatic vasculature, superimpose cell-type-specific and relative deficiencies in S1P production to define ligand source and dose dependence, and correlate receptor engagement to essential functions. In naïve blood vessels, despite broad expression, EC S1PR1 engagement was restricted to resistance-size arteries, lung capillaries, and a subset of high-endothelial venules (HEVs). Similar zonation was observed for albumin extravasation in EC S1PR1-deficient mice, and brain extravasation was reproduced with arterial EC-selective S1pr1 deletion. In lymphatic ECs, S1PR1 engagement was high in collecting vessels and lymph nodes and low in blind-ended capillaries that drain tissue fluids. While EC S1P production sustained S1PR1 signalling in lymphatics and HEV, haematopoietic cells provided ∼90% of plasma S1P and sustained signalling in resistance arteries and lung capillaries. S1PR1 signalling and endothelial function were both surprisingly sensitive to reductions in plasma S1P with apparent saturation around 50% of normal levels. S1PR1 engagement did not depend on sex or age but modestly increased in arteries in hypertension and diabetes. Sphingosine kinase (Sphk)-2 deficiency also increased S1PR1 engagement selectively in arteries, which could be attributed to Sphk1-dependent S1P release from perivascular macrophages. CONCLUSION This study highlights vessel subtype-specific S1PR1 functions and mechanisms of engagement and supports the relevance of S1P as circulating biomarker for endothelial function.
Collapse
Affiliation(s)
- Ilaria Del Gaudio
- Université Paris Cité, Paris Cardiovascular Research Centre, INSERM U970, 56 Rue Leblanc, F-75015 Paris, France
| | - Anja Nitzsche
- Université Paris Cité, Paris Cardiovascular Research Centre, INSERM U970, 56 Rue Leblanc, F-75015 Paris, France
| | - Kevin Boyé
- Université Paris Cité, Paris Cardiovascular Research Centre, INSERM U970, 56 Rue Leblanc, F-75015 Paris, France
| | - Philippe Bonnin
- Physiologie Clinique, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Lariboisière, Paris, France
- Université Paris Cité, INSERM U1144, UFR de Pharmacie, Paris, France
| | - Mathilde Poulet
- Université Paris Cité, Paris Cardiovascular Research Centre, INSERM U970, 56 Rue Leblanc, F-75015 Paris, France
| | - Toan Q Nguyen
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| | - Ludovic Couty
- Université Paris Cité, Paris Cardiovascular Research Centre, INSERM U970, 56 Rue Leblanc, F-75015 Paris, France
| | - Hoa T T Ha
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| | - Dat T Nguyen
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| | - Amaury Cazenave-Gassiot
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| | - Khaoula Ben Alaya
- Université Paris Cité, Paris Cardiovascular Research Centre, INSERM U970, 56 Rue Leblanc, F-75015 Paris, France
| | - Patrice Thérond
- Service de Biochimie, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital de Bicêtre, Le Kremlin Bicêtre, France
- UFR de Pharmacie, EA 4529, Châtenay-Malabry, France
| | - Jerold Chun
- Neuroscience Drug Discovery, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Markus R Wenk
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| | - Richard L Proia
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Institutes of Health, Bethesda, MD, USA
| | - Daniel Henrion
- MitoVasc Department, Angers University, Team 2 (CarMe), Angers University Hospital (CHU of Angers), CNRS, INSERM U1083, Angers, France
| | - Long N Nguyen
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| | - Anne Eichmann
- Université Paris Cité, Paris Cardiovascular Research Centre, INSERM U970, 56 Rue Leblanc, F-75015 Paris, France
- Department of Internal Medicine and Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, USA
| | - Eric Camerer
- Université Paris Cité, Paris Cardiovascular Research Centre, INSERM U970, 56 Rue Leblanc, F-75015 Paris, France
| |
Collapse
|
8
|
Yang L, Han Y, Zhang T, Dong X, Ge J, Roy A, Zhu J, Lu T, Jeya Vandana J, de Silva N, Robertson CC, Xiang JZ, Pan C, Sun Y, Que J, Evans T, Liu C, Wang W, Naji A, Parker SCJ, Schwartz RE, Chen S. Human vascularized macrophage-islet organoids to model immune-mediated pancreatic β cell pyroptosis upon viral infection. Cell Stem Cell 2024; 31:1612-1629.e8. [PMID: 39232561 PMCID: PMC11546835 DOI: 10.1016/j.stem.2024.08.007] [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: 11/17/2023] [Revised: 06/05/2024] [Accepted: 08/09/2024] [Indexed: 09/06/2024]
Abstract
There is a paucity of human models to study immune-mediated host damage. Here, we utilized the GeoMx spatial multi-omics platform to analyze immune cell changes in COVID-19 pancreatic autopsy samples, revealing an accumulation of proinflammatory macrophages. Single-cell RNA sequencing (scRNA-seq) analysis of human islets exposed to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) or coxsackievirus B4 (CVB4) viruses identified activation of proinflammatory macrophages and β cell pyroptosis. To distinguish viral versus proinflammatory-macrophage-mediated β cell pyroptosis, we developed human pluripotent stem cell (hPSC)-derived vascularized macrophage-islet (VMI) organoids. VMI organoids exhibited enhanced marker expression and function in both β cells and endothelial cells compared with separately cultured cells. Notably, proinflammatory macrophages within VMI organoids induced β cell pyroptosis. Mechanistic investigations highlighted TNFSF12-TNFRSF12A involvement in proinflammatory-macrophage-mediated β cell pyroptosis. This study established hPSC-derived VMI organoids as a valuable tool for studying immune-cell-mediated host damage and uncovered the mechanism of β cell damage during viral exposure.
Collapse
Affiliation(s)
- Liuliu Yang
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; Center for Genomic Health, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Disease, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China; Tianjin Institute of Health Science, Tianjin 301600, China.
| | - Yuling Han
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; Center for Genomic Health, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Tuo Zhang
- Genomic Resource Core Facility, Weill Cornell Medicine, New York, NY 10065, USA
| | - Xue Dong
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Jian Ge
- Columbia Center for Human Development, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Aadita Roy
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Jiajun Zhu
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; Center for Genomic Health, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Tiankun Lu
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; Center for Genomic Health, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - J Jeya Vandana
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; Center for Genomic Health, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Neranjan de Silva
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; Center for Genomic Health, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Catherine C Robertson
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Jenny Z Xiang
- Genomic Resource Core Facility, Weill Cornell Medicine, New York, NY 10065, USA
| | - Chendong Pan
- Genomic Resource Core Facility, Weill Cornell Medicine, New York, NY 10065, USA
| | - Yanjie Sun
- Genomic Resource Core Facility, Weill Cornell Medicine, New York, NY 10065, USA
| | - Jianwen Que
- Columbia Center for Human Development, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Todd Evans
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; Center for Genomic Health, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Chengyang Liu
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Wei Wang
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Ali Naji
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Stephen C J Parker
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA; Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA; Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Robert E Schwartz
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA.
| | - Shuibing Chen
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; Center for Genomic Health, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
9
|
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
|
10
|
Kim JD, Jain A, Fang L. Mitigating Vascular Inflammation by Mimicking AIBP Mechanisms: A New Therapeutic End for Atherosclerotic Cardiovascular Disease. Int J Mol Sci 2024; 25:10314. [PMID: 39408645 PMCID: PMC11477018 DOI: 10.3390/ijms251910314] [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: 08/14/2024] [Revised: 09/10/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024] Open
Abstract
Atherosclerosis, characterized by the accumulation of lipoproteins and lipids within the vascular wall, underlies a heart attack, stroke, and peripheral artery disease. Endothelial inflammation is the primary component driving atherosclerosis, promoting leukocyte adhesion molecule expression (e.g., E-selectin), inducing chemokine secretion, reducing the production of nitric oxide (NO), and enhancing the thrombogenic potential. While current therapies, such as statins, colchicine, anti-IL1β, and sodium-glucose cotransporter 2 (SGLT2) inhibitors, target systemic inflammation, none of them addresses endothelial cell (EC) inflammation, a critical contributor to disease progression. Targeting endothelial inflammation is clinically significant because it can mitigate the root cause of atherosclerosis, potentially preventing disease progression, while reducing the side effects associated with broader anti-inflammatory treatments. Recent studies highlight the potential of the APOA1 binding protein (AIBP) to reduce systemic inflammation in mice. Furthermore, its mechanism of action also guides the design of a potential targeted therapy against a particular inflammatory signaling pathway. This review discusses the unique advantages of repressing vascular inflammation or enhancing vascular quiescence and the associated benefits of reducing thrombosis. This approach offers a promising avenue for more effective and targeted interventions to improve patient outcomes.
Collapse
Affiliation(s)
- Jun-Dae Kim
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Abhishek Jain
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, USA;
| | - Longhou Fang
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX 77030, USA
- Weill Cornell Medical College, Cornell University, Ithaca, NY 14850, USA
| |
Collapse
|
11
|
Alshaikh RA, Chullipalliyalil K, Waeber C, Ryan KB. Extended siponimod release via low-porosity PLGA fibres: a comprehensive three-month in vitro evaluation for neovascular ocular diseases. Biomater Sci 2024; 12:4823-4844. [PMID: 39157879 DOI: 10.1039/d4bm00339j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/20/2024]
Abstract
Neovascular ocular diseases are among the most common causes of preventable or treatable vision loss. Their management involves lifelong, intravitreal injections of anti-vascular endothelial growth factor (VEGF) therapeutics to inhibit neovascularization, the key pathological step in these diseases. Anti-VEGF products approved for ocular administration are expensive biological agents with limited stability and short half-life. Additionally, their therapeutic advantages are hindered by high treatment resistance, poor patient compliance and the need for frequent, invasive administration. Herein, we used electrospinning to develop a unique, non-porous, PLGA implant for the ocular delivery of siponimod to improve ocular neovascular disease management. Siponimod is an FDA-approved drug for multiple sclerosis with a novel indication as a potential ocular angiogenesis inhibitor. The electrospinning conditions were optimised to produce a microfibrous, PLGA matte that was cut and rolled into the desired implant size. Physical characterisation techniques (Raman, PXRD, DSC and FTIR) indicated siponimod was distributed uniformly within the electrospun fibres as a stabilised, amorphous, solid dispersion with a character modifying drug-polymer interaction. Siponimod dispersion and drug-polymer interactions contributed to the formation of smooth fibres, with reduced porous structures. The apparent reduced porosity, coupled with the drug's hydrophobic dispersion, afforded resistance to water penetration. This led to a slow, controlled, Higuchi-type drug diffusion, with ∼30% of the siponimod load released over 90 days. The released drug inhibited human retinal microvascular endothelial cell migration and did not affect the cells' metabolic activity at different time points. The electrospun implant was physically stable after incubation under stress conditions for three months. This novel siponimod intravitreal implant broadens the therapeutic possibilities for neovascular ocular diseases, representing a potential alternative to biological, anti-VEGF treatments due to lower financial and stability burdens. Additionally, siponimod interaction with PLGA provides a unique opportunity to sustain the drug release from the electrospun fibres, thereby reducing the frequency of intravitreal injection and improving patient adherence.
Collapse
Affiliation(s)
- Rasha A Alshaikh
- School of Pharmacy, University College Cork, Cork, Ireland.
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | | | - Christian Waeber
- School of Pharmacy, University College Cork, Cork, Ireland.
- Department of Pharmacology and Therapeutics, School of Medicine, University College Cork, Cork, Ireland
| | - Katie B Ryan
- School of Pharmacy, University College Cork, Cork, Ireland.
- SSPC The SFI Research Centre for Pharmaceuticals, School of Pharmacy, University College Cork, Cork, Ireland
| |
Collapse
|
12
|
Yang L, Han Y, Zhang T, Dong X, Ge J, Roy A, Zhu J, Lu T, Vandana JJ, de Silva N, Robertson CC, Xiang JZ, Pan C, Sun Y, Que J, Evans T, Liu C, Wang W, Naji A, Parker SC, Schwartz RE, Chen S. Human Vascularized Macrophage-Islet Organoids to Model Immune-Mediated Pancreatic β cell Pyroptosis upon Viral Infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.05.606734. [PMID: 39149298 PMCID: PMC11326194 DOI: 10.1101/2024.08.05.606734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
There is a paucity of human models to study immune-mediated host damage. Here, we utilized the GeoMx spatial multi-omics platform to analyze immune cell changes in COVID-19 pancreatic autopsy samples, revealing an accumulation of proinflammatory macrophages. Single cell RNA-seq analysis of human islets exposed to SARS-CoV-2 or Coxsackievirus B4 (CVB4) viruses identified activation of proinflammatory macrophages and β cell pyroptosis. To distinguish viral versus proinflammatory macrophage-mediated β cell pyroptosis, we developed human pluripotent stem cell (hPSC)-derived vascularized macrophage-islet (VMI) organoids. VMI organoids exhibited enhanced marker expression and function in both β cells and endothelial cells compared to separately cultured cells. Notably, proinflammatory macrophages within VMI organoids induced β cell pyroptosis. Mechanistic investigations highlighted TNFSF12-TNFRSF12A involvement in proinflammatory macrophage-mediated β cell pyroptosis. This study established hPSC-derived VMI organoids as a valuable tool for studying immune cell-mediated host damage and uncovered mechanism of β cell damage during viral exposure.
Collapse
Affiliation(s)
- Liuliu Yang
- Department of Surgery, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
- Center for Genomic Health, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Disease, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
- Tianjin Institute of Health Science, Tianjin 301600, China
| | - Yuling Han
- Department of Surgery, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
- Center for Genomic Health, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Tuo Zhang
- Genomic Resource Core Facility, Weill Cornell Medicine, New York, NY 10065, USA
| | - Xue Dong
- Department of Surgery, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
| | - Jian Ge
- Columbia Center for Human Development, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Aadita Roy
- Department of Surgery, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
| | - Jiajun Zhu
- Department of Surgery, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
- Center for Genomic Health, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
| | - Tiankun Lu
- Department of Surgery, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
- Center for Genomic Health, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
| | - J. Jeya Vandana
- Department of Surgery, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
- Center for Genomic Health, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
| | - Neranjan de Silva
- Department of Surgery, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
- Center for Genomic Health, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
| | - Catherine C. Robertson
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Jenny Z Xiang
- Genomic Resource Core Facility, Weill Cornell Medicine, New York, NY 10065, USA
| | - Chendong Pan
- Genomic Resource Core Facility, Weill Cornell Medicine, New York, NY 10065, USA
| | - Yanjie Sun
- Genomic Resource Core Facility, Weill Cornell Medicine, New York, NY 10065, USA
| | - Jianwen Que
- Columbia Center for Human Development, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Todd Evans
- Department of Surgery, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
- Center for Genomic Health, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
| | - Chengyang Liu
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Wei Wang
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Ali Naji
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Stephen C.J. Parker
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Robert E. Schwartz
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA. New York 10021, USA
| | - Shuibing Chen
- Department of Surgery, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
- Center for Genomic Health, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
| |
Collapse
|
13
|
Sheng X, Zhang C, Zhao J, Xu J, Zhang P, Ding Q, Zhang J. Microvascular destabilization and intricated network of the cytokines in diabetic retinopathy: from the perspective of cellular and molecular components. Cell Biosci 2024; 14:85. [PMID: 38937783 PMCID: PMC11212265 DOI: 10.1186/s13578-024-01269-7] [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: 04/18/2024] [Accepted: 06/19/2024] [Indexed: 06/29/2024] Open
Abstract
Microvascular destabilization is the primary cause of the inner blood-retinal barrier (iBRB) breakdown and increased vascular leakage in diabetic retinopathy (DR). Microvascular destabilization results from the combinational effects of increased levels of growth factors and cytokines, involvement of inflammation, and the changed cell-to-cell interactions, especially the loss of endothelial cells and pericytes, due to hyperglycemia and hypoxia. As the manifestation of microvascular destabilization, the fluid transports via paracellular and transcellular routes increase due to the disruption of endothelial intercellular junctional complexes and/or the altered caveolar transcellular transport across the retinal vascular endothelium. With diabetes progression, the functional and the structural changes of the iBRB components, including the cellular and noncellular components, further facilitate and aggravate microvascular destabilization, resulting in macular edema, the neuroretinal damage and the dysfunction of retinal inner neurovascular unit (iNVU). Although there have been considerable recent advances towards a better understanding of the complex cellular and molecular network underlying the microvascular destabilization, some still remain to be fully elucidated. Recent data indicate that targeting the intricate signaling pathways may allow to against the microvascular destabilization. Therefore, efforts have been made to better clarify the cellular and molecular mechanisms that are involved in the microvascular destabilization in DR. In this review, we discuss: (1) the brief introduction of DR and microvascular destabilization; (2) the cellular and molecular components of iBRB and iNVU, and the breakdown of iBRB; (3) the matrix and cell-to-cell contacts to maintain microvascular stabilization, including the endothelial glycocalyx, basement membrane, and various cell-cell interactions; (4) the molecular mechanisms mediated cell-cell contacts and vascular cell death; (5) the altered cytokines and signaling pathways as well as the intricate network of the cytokines involved in microvascular destabilization. This comprehensive review aimed to provide the insights for microvascular destabilization by targeting the key molecules or specific iBRB cells, thus restoring the function and structure of iBRB and iNVU, to treat DR.
Collapse
Affiliation(s)
- Xia Sheng
- People's Hospital of Huangdao District, Qingdao, Shandong Province, China
| | - Chunmei Zhang
- People's Hospital of Huangdao District, Qingdao, Shandong Province, China
| | - Jiwei Zhao
- People's Hospital of Huangdao District, Qingdao, Shandong Province, China
| | - Jianping Xu
- People's Hospital of Huangdao District, Qingdao, Shandong Province, China.
| | - Peng Zhang
- People's Hospital of Huangdao District, Qingdao, Shandong Province, China.
| | - Quanju Ding
- People's Hospital of Huangdao District, Qingdao, Shandong Province, China.
| | - Jingfa Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, National Clinical Research Center for Eye Diseases, Shanghai, China.
- The International Eye Research Institute of The Chinese University of Hong Kong (Shenzhen), Shenzhen, China.
- C-MER (Shenzhen) Dennis Lam Eye Hospital, Shenzhen, China.
- C-MER International Eye Care Group, C-MER Dennis Lam & Partners Eye Center, Hong Kong, China.
| |
Collapse
|
14
|
Raza Q, Nadeem T, Youn SW, Swaminathan B, Gupta A, Sargis T, Du J, Cuervo H, Eichmann A, Ackerman SL, Naiche LA, Kitajewski J. Notch signaling regulates UNC5B to suppress endothelial proliferation, migration, junction activity, and retinal plexus branching. Sci Rep 2024; 14:13603. [PMID: 38866944 PMCID: PMC11169293 DOI: 10.1038/s41598-024-64375-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 06/07/2024] [Indexed: 06/14/2024] Open
Abstract
Notch signaling guides vascular development and function by regulating diverse endothelial cell behaviors, including migration, proliferation, vascular density, endothelial junctions, and polarization in response to flow. Notch proteins form transcriptional activation complexes that regulate endothelial gene expression, but few of the downstream effectors that enable these phenotypic changes have been characterized in endothelial cells, limiting our understanding of vascular Notch activities. Using an unbiased screen of translated mRNA rapidly regulated by Notch signaling, we identified novel in vivo targets of Notch signaling in neonatal mouse brain endothelium, including UNC5B, a member of the netrin family of angiogenic-regulatory receptors. Endothelial Notch signaling rapidly upregulates UNC5B in multiple endothelial cell types. Loss or gain of UNC5B recapitulated specific Notch-regulated phenotypes. UNC5B expression inhibited endothelial migration and proliferation and was required for stabilization of endothelial junctions in response to shear stress. Loss of UNC5B partially or wholly blocked the ability of Notch activation to regulate these endothelial cell behaviors. In the developing mouse retina, endothelial-specific loss of UNC5B led to excessive vascularization, including increased vascular outgrowth, density, and branchpoint count. These data indicate that Notch signaling upregulates UNC5B as an effector protein to control specific endothelial cell behaviors and inhibit angiogenic growth.
Collapse
Affiliation(s)
- Qanber Raza
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, 1853 W Polk St, Rm 522 (MC 901), Chicago, IL, 60612, USA
| | - Taliha Nadeem
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, 1853 W Polk St, Rm 522 (MC 901), Chicago, IL, 60612, USA
| | - Seock-Won Youn
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, 1853 W Polk St, Rm 522 (MC 901), Chicago, IL, 60612, USA
| | - Bhairavi Swaminathan
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, 1853 W Polk St, Rm 522 (MC 901), Chicago, IL, 60612, USA
| | - Ahana Gupta
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, 1853 W Polk St, Rm 522 (MC 901), Chicago, IL, 60612, USA
| | - Timothy Sargis
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, 1853 W Polk St, Rm 522 (MC 901), Chicago, IL, 60612, USA
| | - Jing Du
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, 1853 W Polk St, Rm 522 (MC 901), Chicago, IL, 60612, USA
| | - Henar Cuervo
- Centro Nacional de Investigaciones Cardiovasculares Carlos III- CNIC- (F.S.P), Madrid, Spain
| | | | | | - L A Naiche
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, 1853 W Polk St, Rm 522 (MC 901), Chicago, IL, 60612, USA.
| | - Jan Kitajewski
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, 1853 W Polk St, Rm 522 (MC 901), Chicago, IL, 60612, USA
- University of Illinois Cancer Center, Chicago, USA
| |
Collapse
|
15
|
Nakamura S, Yamamoto R, Matsuda T, Yasuda H, Nishinaka A, Takahashi K, Inoue Y, Kuromitsu S, Shimazawa M, Goto M, Narumiya S, Hara H. Sphingosine-1-phosphate receptor 1/5 selective agonist alleviates ocular vascular pathologies. Sci Rep 2024; 14:9700. [PMID: 38678148 PMCID: PMC11055896 DOI: 10.1038/s41598-024-60540-6] [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: 10/12/2023] [Accepted: 04/24/2024] [Indexed: 04/29/2024] Open
Abstract
Ocular abnormal angiogenesis and edema are featured in several ocular diseases. S1P signaling via S1P1 likely is part of the negative feedback mechanism necessary to maintain vascular health. In this study, we conducted pharmacological experiments to determine whether ASP4058, a sphingosine 1-phosphate receptor 1/5 (S1P1/5) agonist, is useful in abnormal vascular pathology in the eye. First, human retinal microvascular endothelial cells (HRMECs) were examined using vascular endothelial growth factor (VEGF)-induced cell proliferation and hyperpermeability. ASP4058 showed high affinity and inhibited VEGF-induced proliferation and hyperpermeability of HRMECs. Furthermore, S1P1 expression and localization changes were examined in the murine laser-induced choroidal neovascularization (CNV) model, a mouse model of exudative age-related macular degeneration, and the efficacy of ASP4058 was verified. In the CNV model mice, S1P1 tended to decrease in expression immediately after laser irradiation and colocalized with endothelial cells and Müller glial cells. Oral administration of ASP4058 also suppressed vascular hyperpermeability and CNV, and the effect was comparable to that of the intravitreal administration of aflibercept, an anti-VEGF drug. Next, efficacy was also examined in a retinal vein occlusion (RVO) model in which retinal vascular permeability was increased. ASP4058 dose-dependently suppressed the intraretinal edema. In addition, it suppressed the expansion of the perfusion area observed in the RVO model. ASP4058 also suppressed the production of VEGF in the eye. Collectively, ASP4058 can be a potential therapeutic agent that normalizes abnormal vascular pathology, such as age-related macular degeneration and RVO, through its direct action on endothelial cells.
Collapse
Affiliation(s)
- Shinsuke Nakamura
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, 501-1196, Japan
| | - Rie Yamamoto
- Discovery Accelerator, Astellas Pharma Inc., Tsukuba, Japan
- Alliance Laboratory for Advanced Medical Research, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takaya Matsuda
- Pharmaceutical Research and Technology Labs, Astellas Pharma Inc., Yaizu, Japan
| | - Hiroto Yasuda
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, 501-1196, Japan
| | - Anri Nishinaka
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, 501-1196, Japan
| | - Kei Takahashi
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, 501-1196, Japan
| | - Yuki Inoue
- Astellas Institute for Regenerative Medicine, Marlborough, MA, USA
| | - Sadao Kuromitsu
- Discovery Accelerator, Astellas Pharma Inc., Tsukuba, Japan
- Alliance Laboratory for Advanced Medical Research, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masamitsu Shimazawa
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, 501-1196, Japan
| | - Masahide Goto
- Astellas Institute for Regenerative Medicine, Marlborough, MA, USA
| | - Shuh Narumiya
- Department of Drug Discovery Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hideaki Hara
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, 501-1196, Japan.
| |
Collapse
|
16
|
Wille A, Weske S, von Wnuck Lipinski K, Wollnitzke P, Schröder NH, Thomas N, Nowak MK, Deister-Jonas J, Behr B, Keul P, Levkau B. Sphingosine-1-phosphate promotes osteogenesis by stimulating osteoblast growth and neovascularization in a vascular endothelial growth factor-dependent manner. J Bone Miner Res 2024; 39:357-372. [PMID: 38477738 PMCID: PMC11240155 DOI: 10.1093/jbmr/zjae006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 12/19/2023] [Accepted: 12/29/2023] [Indexed: 03/14/2024]
Abstract
Sphingosine-1-phosphate (S1P) plays multiple roles in bone metabolism and regeneration. Here, we have identified a novel S1P-regulated osteoanabolic mechanism functionally connecting osteoblasts (OBs) to the highly specialized bone vasculature. We demonstrate that S1P/S1PR3 signaling in OBs stimulates vascular endothelial growth factor a (VEGFa) expression and secretion to promote bone growth in an autocrine and boost osteogenic H-type differentiation of bone marrow endothelial cells in a paracrine manner. VEGFa-neutralizing antibodies and VEGF receptor inhibition by axitinib abrogated OB growth in vitro and bone formation in male C57BL/6J in vivo following S1P stimulation and S1P lyase inhibition, respectively. Pharmacological S1PR3 inhibition and genetic S1PR3 deficiency suppressed VEGFa production, OB growth in vitro, and inhibited H-type angiogenesis and bone growth in male mice in vivo. Together with previous work on the osteoanabolic functions of S1PR2 and S1PR3, our data suggest that S1P-dependent bone regeneration employs several nonredundant positive feedback loops between OBs and the bone vasculature. The identification of this yet unappreciated aspect of osteoanabolic S1P signaling may have implications for regular bone homeostasis as well as diseases where the bone microvasculature is affected such as age-related osteopenia and posttraumatic bone regeneration.
Collapse
Affiliation(s)
- Annalena Wille
- Institute of Molecular Medicine III, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Sarah Weske
- Institute of Molecular Medicine III, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Karin von Wnuck Lipinski
- Institute of Molecular Medicine III, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Philipp Wollnitzke
- Institute of Molecular Medicine III, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Nathalie H Schröder
- Institute of Molecular Medicine III, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Nadine Thomas
- Institute of Molecular Medicine III, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Melissa K Nowak
- Institute of Molecular Medicine III, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Jennifer Deister-Jonas
- Institute of Molecular Medicine III, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Björn Behr
- Department of Plastic Surgery, University Hospital BG Bergmannsheil, 44789 Bochum, Germany
| | - Petra Keul
- Institute of Molecular Medicine III, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Bodo Levkau
- Institute of Molecular Medicine III, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| |
Collapse
|
17
|
Guo Y, Zhang S, Wang D, Heng BC, Deng X. Role of cell rearrangement and related signaling pathways in the dynamic process of tip cell selection. Cell Commun Signal 2024; 22:24. [PMID: 38195565 PMCID: PMC10777628 DOI: 10.1186/s12964-023-01364-1] [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: 06/14/2023] [Accepted: 10/25/2023] [Indexed: 01/11/2024] Open
Abstract
Angiogenesis is a complex, highly-coordinated and multi-step process of new blood vessel formation from pre-existing blood vessels. When initiated, the sprouting process is spearheaded by the specialized endothelial cells (ECs) known as tip cells, which guide the organization of accompanying stalk cells and determine the function and morphology of the finally-formed blood vessels. Recent studies indicate that the orchestration and coordination of angiogenesis involve dynamic tip cell selection, which is the competitive selection of cells to lead the angiogenic sprouts. Therefore, this review attempt to summarize the underlying mechanisms involved in tip cell specification in a dynamic manner to enable readers to gain a systemic and overall understanding of tip cell formation, involving cooperative interaction of cell rearrangement with Notch and YAP/TAZ signaling. Various mechanical and chemical signaling cues are integrated to ensure the right number of cells at the right place during angiogenesis, thereby precisely orchestrating morphogenic functions that ensure correct patterning of blood vessels. Video Abstract.
Collapse
Affiliation(s)
- Yaru Guo
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Shihan Zhang
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Dandan Wang
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Boon Chin Heng
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, China.
- NMPA Key Laboratory for Dental Materials, Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, 100081, China.
| | - Xuliang Deng
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China.
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China.
- Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, China.
| |
Collapse
|
18
|
Kossack ME, Tian L, Bowie K, Plavicki JS. Defining the cellular complexity of the zebrafish bipotential gonad. Biol Reprod 2023; 109:586-600. [PMID: 37561446 PMCID: PMC10651076 DOI: 10.1093/biolre/ioad096] [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] [Indexed: 08/11/2023] Open
Abstract
Zebrafish are routinely used to model reproductive development, function, and disease, yet we still lack a clear understanding of the fundamental steps that occur during early bipotential gonad development, including when endothelial cells, pericytes, and macrophage arrive at the bipotential gonad to support gonad growth and differentiation. Here, we use a combination of transgenic reporters and single-cell sequencing analyses to define the arrival of different critical cell types to the larval zebrafish gonad. We determined that blood initially reaches the gonad via a vessel formed from the swim bladder artery, which we have termed the gonadal artery. We find that vascular and lymphatic development occurs concurrently in the bipotential zebrafish gonad and our data suggest that similar to what has been observed in developing zebrafish embryos, lymphatic endothelial cells in the gonad may be derived from vascular endothelial cells. We mined preexisting sequencing datasets to determine whether ovarian pericytes had unique gene expression signatures. We identified 215 genes that were uniquely expressed in ovarian pericytes, but not expressed in larval pericytes. Similar to what has been shown in the mouse ovary, our data suggest that pdgfrb+ pericytes may support the migration of endothelial tip cells during ovarian angiogenesis. Using a macrophage-driven photoconvertible protein, we found that macrophage established a nascent resident population as early as 12 dpf and can be observed removing cellular material during gonadal differentiation. This foundational information demonstrates that the early bipotential gonad contains complex cellular interactions, which likely shape the health and function of the mature gonad.
Collapse
Affiliation(s)
- Michelle E Kossack
- Pathology and Laboratory Medicine Department, Brown University, Providence, RI, USA
| | - Lucy Tian
- Pathology and Laboratory Medicine Department, Brown University, Providence, RI, USA
| | - Kealyn Bowie
- Pathology and Laboratory Medicine Department, Brown University, Providence, RI, USA
| | - Jessica S Plavicki
- Pathology and Laboratory Medicine Department, Brown University, Providence, RI, USA
| |
Collapse
|
19
|
Cleuren A, Molema G. Organotypic heterogeneity in microvascular endothelial cell responses in sepsis-a molecular treasure trove and pharmacological Gordian knot. Front Med (Lausanne) 2023; 10:1252021. [PMID: 38020105 PMCID: PMC10665520 DOI: 10.3389/fmed.2023.1252021] [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: 07/03/2023] [Accepted: 10/18/2023] [Indexed: 12/01/2023] Open
Abstract
In the last decades, it has become evident that endothelial cells (ECs) in the microvasculature play an important role in the pathophysiology of sepsis-associated multiple organ dysfunction syndrome (MODS). Studies on how ECs orchestrate leukocyte recruitment, control microvascular integrity and permeability, and regulate the haemostatic balance have provided a wealth of knowledge and potential molecular targets that could be considered for pharmacological intervention in sepsis. Yet, this information has not been translated into effective treatments. As MODS affects specific vascular beds, (organotypic) endothelial heterogeneity may be an important contributing factor to this lack of success. On the other hand, given the involvement of ECs in sepsis, this heterogeneity could also be leveraged for therapeutic gain to target specific sites of the vasculature given its full accessibility to drugs. In this review, we describe current knowledge that defines heterogeneity of organ-specific microvascular ECs at the molecular level and elaborate on studies that have reported EC responses across organ systems in sepsis patients and animal models of sepsis. We discuss hypothesis-driven, single-molecule studies that have formed the basis of our understanding of endothelial cell engagement in sepsis pathophysiology, and include recent studies employing high-throughput technologies. The latter deliver comprehensive data sets to describe molecular signatures for organotypic ECs that could lead to new hypotheses and form the foundation for rational pharmacological intervention and biomarker panel development. Particularly results from single cell RNA sequencing and spatial transcriptomics studies are eagerly awaited as they are expected to unveil the full spatiotemporal signature of EC responses to sepsis. With increasing awareness of the existence of distinct sepsis subphenotypes, and the need to develop new drug regimen and companion diagnostics, a better understanding of the molecular pathways exploited by ECs in sepsis pathophysiology will be a cornerstone to halt the detrimental processes that lead to MODS.
Collapse
Affiliation(s)
- Audrey Cleuren
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Grietje Molema
- Department Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
20
|
Yu B, Shen K, Li T, Li J, Meng M, Liu W, Tang Q, Zhu T, Wang X, Leung SWS, Shi Y. Glycolytic enzyme PFKFB3 regulates sphingosine 1-phosphate receptor 1 in proangiogenic glomerular endothelial cells under diabetic condition. Am J Physiol Cell Physiol 2023; 325:C1354-C1368. [PMID: 37781737 PMCID: PMC10861147 DOI: 10.1152/ajpcell.00261.2023] [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: 06/15/2023] [Revised: 09/21/2023] [Accepted: 09/23/2023] [Indexed: 10/03/2023]
Abstract
Glomerular angiogenesis is a characteristic feature of diabetic nephropathy (DN). Enhanced glycolysis plays a crucial role in angiogenesis. The present study was designed to investigate the role of glycolysis in glomerular endothelial cells (GECs) in a mouse model of DN. Mouse renal cortex and isolated glomerular cells were collected for single-cell and RNA sequencing. Cultured GECs were exposed to high glucose in the presence (proangiogenic) and absence of a vascular sprouting regimen. MicroRNA-590-3p was delivered by lipofectamine in vivo and in vitro. In the present study, a subgroup of GECs with proangiogenic features was identified in diabetic kidneys by using sequencing analyses. In cultured proangiogenic GECs, high glucose increased glycolysis and phosphofructokinase/fructose bisphosphatase 3 (PFKFB3) protein expression, which were inhibited by overexpressing miRNA-590-3p. Mimics of miRNA-590-3p also increased receptor for sphingosine 1-phosphate (S1pR1) expression, an angiogenesis regulator, in proangiogenic GECs challenged with high glucose. Inhibition of PFKFB3 by pharmacological and genetic approaches upregulated S1pR1 protein in vitro. Mimics of miRNA-590-3p significantly reduced migration and angiogenic potential in proangiogenic GECs challenged with high glucose. Ten-week-old type 2 diabetic mice had elevated urinary albumin levels, reduced renal cortex miRNA-590-3p expression, and disarrangement of glomerular endothelial cell fenestration. Overexpressing miRNA-590-3p via perirenal adipose tissue injection restored endothelial cell fenestration and reduced urinary albumin levels in diabetic mice. Therefore, the present study identifies a subgroup of GECs with proangiogenic features in mice with DN. Local administration of miRNA-590-3p mimics reduces glycolytic rate and upregulates S1pR1 protein expression in proangiogenic GECs. The protective effects of miRNA-590-3p provide therapeutic potential in DN treatment.NEW & NOTEWORTHY Proangiogenetic glomerular endothelial cells (GECs) are activated in diabetic nephropathy. High glucose upregulates glycolytic enzyme phosphofructokinase/fructose bisphosphatase 3 (PFKFB3) in proangiogenetic cells. PFKFB3 protects the glomerular filtration barrier by targeting endothelial S1pR1. MiRNA-590-3p restores endothelial cell function and mitigates diabetic nephropathy.
Collapse
Affiliation(s)
- Baixue Yu
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
- Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Kaiyuan Shen
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Tingting Li
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
- Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Jiawei Li
- Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Mei Meng
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
- Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Wenjie Liu
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
- Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Qunye Tang
- Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Tongyu Zhu
- Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Xin Wang
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Susan W S Leung
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, People's Republic of China
| | - Yi Shi
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
- Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
21
|
Mu X, Hu K, Wei A, Bai J, Feng L, Jiang J. TiO 2 nanoparticles promote tumor metastasis by eliciting pro-metastatic extracellular vesicles. J Nanobiotechnology 2023; 21:392. [PMID: 37891598 PMCID: PMC10604521 DOI: 10.1186/s12951-023-02142-4] [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: 07/21/2023] [Accepted: 10/04/2023] [Indexed: 10/29/2023] Open
Abstract
The development of nanotechnology has provided numerous possibilities for the diagnosis and treatment of cancer. Paradoxically, some in vivo experimental studies have also shown that nanoparticles (NPs) could promote tumor progression, but the specific mechanism is not yet clear. Primary tumors can release extracellular vesicles (EVs) which can promote the inoculation and growth of tumor cells that have metastasized to distant organs. So, whether nanomaterials can promote tumor progression through tumor-derived EVs deserves further research. Here, we showed that TiO2 NPs, widely used in nanomedicine, could trigger tumor-derived EVs with enhanced pro-metastatic capacity in vitro and in vivo. Mechanically, miR-301a-3p derived from NPs-elicited EVs could be delivered into vascular endothelial cells, which inhibited the expression of VEGFR2 and VE-cadherin by targeting S1PR1, leading to disrupt the tight junctions of vascular endothelial cells, thus to promote vascular permeability and angiogenesis, and induce the formation of pre-metastasis niches in vivo. This study emphasizes that it is urgent to consider the effect of NPs on EVs under long-term use conditions when designing nanodrugs for cancer treatment.
Collapse
Affiliation(s)
- Xupeng Mu
- Scientifc Research Center, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Kebang Hu
- Department of Urology, Lequn Branch, The First Hospital of Jilin University, Changchun, 130031, China
| | - Anhui Wei
- Department of Regenerative Medicine, College of Pharmacy, Jilin University, Changchun, 130021, China
| | - Jinping Bai
- Department of Chronic Disease, Jilin Province FAW General Hospital, Changchun, 130013, China
| | - Li Feng
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, 130033, China.
| | - Jinlan Jiang
- Scientifc Research Center, China-Japan Union Hospital of Jilin University, Changchun, 130033, China.
| |
Collapse
|
22
|
Kim D, Tian W, Wu TTH, Xiang M, Vinh R, Chang JL, Gu S, Lee S, Zhu Y, Guan T, Schneider EC, Bao E, Dixon JB, Kao P, Pan J, Rockson SG, Jiang X, Nicolls MR. Abnormal Lymphatic Sphingosine-1-Phosphate Signaling Aggravates Lymphatic Dysfunction and Tissue Inflammation. Circulation 2023; 148:1231-1249. [PMID: 37609838 PMCID: PMC10592179 DOI: 10.1161/circulationaha.123.064181] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 07/31/2023] [Indexed: 08/24/2023]
Abstract
BACKGROUND Lymphedema is a global health problem with no effective drug treatment. Enhanced T-cell immunity and abnormal lymphatic endothelial cell (LEC) signaling are promising therapeutic targets for this condition. Sphingosine-1-phosphate (S1P) mediates a key signaling pathway required for normal LEC function, and altered S1P signaling in LECs could lead to lymphatic disease and pathogenic T-cell activation. Characterizing this biology is relevant for developing much needed therapies. METHODS Human and mouse lymphedema was studied. Lymphedema was induced in mice by surgically ligating the tail lymphatics. Lymphedematous dermal tissue was assessed for S1P signaling. To verify the role of altered S1P signaling effects in lymphatic cells, LEC-specific S1pr1-deficient (S1pr1LECKO) mice were generated. Disease progression was quantified by tail-volumetric and -histopathologic measurements over time. LECs from mice and humans, with S1P signaling inhibition, were then cocultured with CD4 T cells, followed by an analysis of CD4 T-cell activation and pathway signaling. Last, animals were treated with a monoclonal antibody specific to P-selectin to assess its efficacy in reducing lymphedema and T-cell activation. RESULTS Human and experimental lymphedema tissues exhibited decreased LEC S1P signaling through S1P receptor 1 (S1PR1). LEC S1pr1 loss-of-function exacerbated lymphatic vascular insufficiency, tail swelling, and increased CD4 T-cell infiltration in mouse lymphedema. LECs, isolated from S1pr1LECKO mice and cocultured with CD4 T cells, resulted in augmented lymphocyte differentiation. Inhibiting S1PR1 signaling in human dermal LECs promoted T-helper type 1 and 2 (Th1 and Th2) cell differentiation through direct cell contact with lymphocytes. Human dermal LECs with dampened S1P signaling exhibited enhanced P-selectin, an important cell adhesion molecule expressed on activated vascular cells. In vitro, P-selectin blockade reduced the activation and differentiation of Th cells cocultured with shS1PR1-treated human dermal LECs. P-selectin-directed antibody treatment improved tail swelling and reduced Th1/Th2 immune responses in mouse lymphedema. CONCLUSIONS This study suggests that reduction of the LEC S1P signaling aggravates lymphedema by enhancing LEC adhesion and amplifying pathogenic CD4 T-cell responses. P-selectin inhibitors are suggested as a possible treatment for this pervasive condition.
Collapse
Affiliation(s)
- Dongeon Kim
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Wen Tian
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Timothy Ting-Hsuan Wu
- Stanford University School of Medicine, Stanford, California, USA
- Department of Biochemistry, Stanford Bio-X, Stanford, California, USA
| | - Menglan Xiang
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Ryan Vinh
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Jason Lon Chang
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Shenbiao Gu
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Seunghee Lee
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Yu Zhu
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Torrey Guan
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Emilie Claire Schneider
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Evan Bao
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | | | - Peter Kao
- Stanford University School of Medicine, Stanford, California, USA
| | - Junliang Pan
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | | | - Xinguo Jiang
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Mark Robert Nicolls
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
23
|
Wendt TS, Gonzales RJ. Ozanimod differentially preserves human cerebrovascular endothelial barrier proteins and attenuates matrix metalloproteinase-9 activity following in vitro acute ischemic injury. Am J Physiol Cell Physiol 2023; 325:C951-C971. [PMID: 37642239 DOI: 10.1152/ajpcell.00342.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 08/31/2023]
Abstract
Endothelial integrity is critical in mitigating a vicious cascade of secondary injuries following acute ischemic stroke (AIS). Matrix metalloproteinase-9 (MMP-9), a contributor to endothelial integrity loss, is elevated during stroke and is associated with worsened stroke outcome. We investigated the FDA-approved selective sphingosine-1-phosphate receptor 1 (S1PR1) ligand, ozanimod, on the regulation/activity of MMP-9 as well as endothelial barrier components [platelet endothelial cell adhesion molecule 1 (PECAM-1), claudin-5, and zonula occludens 1 (ZO-1)] in human brain microvascular endothelial cells (HBMECs) following hypoxia plus glucose deprivation (HGD). We previously reported that S1PR1 activation improves HBMEC integrity; however, mechanisms underlying S1PR1 involvement in endothelial cell barrier integrity have not been clearly elucidated. We hypothesized that ozanimod would attenuate an HGD-induced increase in MMP-9 activity that would concomitantly attenuate the loss of integral barrier components. Male HBMECs were treated with ozanimod or vehicle and exposed to 3 h of normoxia (21% O2) or HGD (1% O2). Immunoblotting, zymography, qRT-PCR, and immunocytochemical labeling techniques assessed processes related to MMP-9 and barrier markers. We observed that HGD acutely increased MMP-9 activity and reduced claudin-5 and PECAM-1 levels, and ozanimod attenuated these responses. In situ analysis, via PROSPER, suggested that attenuation of MMP-9 activity may be a primary factor in maintaining these integral barrier proteins. We also observed that HGD increased intracellular mechanisms associated with augmented MMP-9 activation; however, ozanimod had no effect on these select factors. Thus, we conclude that ozanimod has the potential to attenuate HGD-mediated decreases in HBMEC integrity in part by decreasing MMP-9 activity as well as preserving barrier properties.NEW & NOTEWORTHY We have identified a potential novel mechanism by which ozanimod, a selective sphingosine-1-phosphate receptor 1 (S1PR1) agonist, attenuates hypoxia plus glucose deprivation (HGD)-induced matrix metalloproteinase-9 (MMP-9) activity and disruptions in integral human brain endothelial cell barrier proteins. Our results suggest that ischemic-like injury elicits increased MMP-9 activity and alterations of barrier integrity proteins in human brain microvascular endothelial cells (HBMECs) and that ozanimod via S1PR1 attenuates these HGD-induced responses, adding to its therapeutic potential in cerebrovascular protection during the acute phase of ischemic stroke.
Collapse
Affiliation(s)
- Trevor S Wendt
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, Arizona, United States
| | - Rayna J Gonzales
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, Arizona, United States
| |
Collapse
|
24
|
Chen C, Wang J, Liu C, Hu J, Liu L. Pioneering therapies for post-infarction angiogenesis: Insight into molecular mechanisms and preclinical studies. Biomed Pharmacother 2023; 166:115306. [PMID: 37572633 DOI: 10.1016/j.biopha.2023.115306] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/01/2023] [Accepted: 08/07/2023] [Indexed: 08/14/2023] Open
Abstract
Acute myocardial infarction (MI), despite significant progress in its treatment, remains a leading cause of chronic heart failure and cardiovascular events such as cardiac arrest. Promoting angiogenesis in the myocardial tissue after MI to restore blood flow in the ischemic and hypoxic tissue is considered an effective treatment strategy. The repair of the myocardial tissue post-MI involves a robust angiogenic response, with mechanisms involved including endothelial cell proliferation and migration, capillary growth, changes in the extracellular matrix, and stabilization of pericytes for neovascularization. In this review, we provide a detailed overview of six key pathways in angiogenesis post-MI: the PI3K/Akt/mTOR signaling pathway, the Notch signaling pathway, the Wnt/β-catenin signaling pathway, the Hippo signaling pathway, the Sonic Hedgehog signaling pathway, and the JAK/STAT signaling pathway. We also discuss novel therapeutic approaches targeting these pathways, including drug therapy, gene therapy, protein therapy, cell therapy, and extracellular vesicle therapy. A comprehensive understanding of these key pathways and their targeted therapies will aid in our understanding of the pathological and physiological mechanisms of angiogenesis after MI and the development and application of new treatment strategies.
Collapse
Affiliation(s)
- Cong Chen
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Jie Wang
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China.
| | - Chao Liu
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Jun Hu
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Lanchun Liu
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| |
Collapse
|
25
|
Karam M, Auclair C. Sphingosine-1-Phosphate as Lung and Cardiac Vasculature Protecting Agent in SARS-CoV-2 Infection. Int J Mol Sci 2023; 24:13088. [PMID: 37685894 PMCID: PMC10488186 DOI: 10.3390/ijms241713088] [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: 06/25/2023] [Revised: 08/16/2023] [Accepted: 08/19/2023] [Indexed: 09/10/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) may cause severe respiratory illness with high mortality. SARS-CoV-2 infection results in a massive inflammatory cell infiltration into the infected lungs accompanied by excessive pro-inflammatory cytokine production. The lung histology of dead patients shows that some areas are severely emphysematous, with enormously dilated blood vessels and micro-thromboses. The inappropriate inflammatory response damaging the pulmonary interstitial arteriolar walls suggests that the respiratory distress may come in a large part from lung vasculature injuries. It has been recently observed that low plasmatic sphingosine-1-phosphate (S1P) is a marker of a worse prognosis of clinical outcome in severe coronavirus disease (COVID) patients. S1P is an angiogenic molecule displaying anti-inflammatory and anti-apoptotic properties, that promote intercellular interactions between endothelial cells and pericytes resulting in the stabilization of arteries and capillaries. In this context, it can be hypothesized that the benefit of a normal S1P level is due to its protective effect on lung vasculature functionality. This paper provides evidence supporting this concept, opening the way for the design of a pharmacological approach involving the use of an S1P lyase inhibitor to increase the S1P level that in turn will rescue the lung vasculature functionality.
Collapse
Affiliation(s)
| | - Christian Auclair
- AC BioTech, Villejuif Biopark, Cancer Campus, 1 mail du Professeur Georges Mathé, 94800 Villejuif, France;
| |
Collapse
|
26
|
Kossack ME, Tian L, Bowie K, Plavicki JS. Defining the cellular complexity of the zebrafish bipotential gonad. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.18.524593. [PMID: 36712047 PMCID: PMC9882255 DOI: 10.1101/2023.01.18.524593] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Zebrafish are routinely used to model reproductive development, function, and disease, yet we still lack a clear understanding of the fundamental steps that occur during early bipotential gonad development, including when endothelial cells, pericytes, and macrophage cells arrive at the bipotential gonad to support gonad growth and differentiation. Here, we use a combination of transgenic reporters and single-cell sequencing analyses to define the arrival of different critical cell types to the larval zebrafish gonad. We determined that blood initially reaches the gonad via a vessel formed from the swim bladder artery, which we have termed the gonadal artery. We find that vascular and lymphatic development occurs concurrently in the bipotential zebrafish gonad and our data suggest that similar to what has been observed in developing zebrafish embryos, lymphatic endothelial cells in the gonad may be derived from vascular endothelial cells. We mined preexisting sequencing data sets to determine whether ovarian pericytes had unique gene expression signatures. We identified 215 genes that were uniquely expressed in ovarian pericytes that were not expressed in larval pericytes. Similar to what has been shown in the mouse ovary, our data suggest that pdgfrb+ pericytes may support the migration of endothelial tip cells during ovarian angiogenesis. Using a macrophage-driven photoconvertible protein, we found that macrophage established a nascent resident population as early as 12 dpf and can be observed removing cellular material during gonadal differentiation. This foundational information demonstrates that the early bipotential gonad contains complex cellular interactions, which likely shape the health and function of the mature, differentiated gonad.
Collapse
|
27
|
Nan W, He Y, Wang S, Zhang Y. Molecular mechanism of VE-cadherin in regulating endothelial cell behaviour during angiogenesis. Front Physiol 2023; 14:1234104. [PMID: 37601629 PMCID: PMC10433914 DOI: 10.3389/fphys.2023.1234104] [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: 06/03/2023] [Accepted: 07/18/2023] [Indexed: 08/22/2023] Open
Abstract
Vascular endothelial (VE)-cadherin, an endothelium-specific adhesion protein, is found in the junctions between endothelial cells (ECs). It's crucial to maintain the homogeneity of ECs. Keeping and controlling the contact between ECs is essential. In addition to its adhesive function, VE-cadherin plays important roles in vascular development, permeability, and tumour angiogenesis. Signal transfer, cytoskeletal reconstruction, and contractile integrating, which are crucial for constructing and maintaining monolayer integrity as well as for repair and regeneration, are the foundation of endothelial cell (EC) junctional dynamics. The molecular basis of adhesion junctions (AJs), which are closely related and work with actin filaments, is provided by the VE-cadherin-catenin complex. They can activate intracellular signals that drive ECs to react or communicate structural changes to junctions. An increasing number of molecules, including the vascular endothelial growth factor receptor 2 (VEGFR2) and vascular endothelial protein tyrosine phosphatase (VE-PTP), have been connected to VE-cadherin in addition to the conventional VE-cadherin-catenin complex. This review demonstrates significant progress in our understanding of the molecular mechanisms that affect VE-cadherin's function in the regulation of EC behaviour during angiogenesis. The knowledge of the molecular processes that control VE-cadherin's role in the regulation of EC behaviour during angiogenesis has recently advanced, as shown in this review.
Collapse
Affiliation(s)
- Weijin Nan
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
| | - Yuxi He
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
| | - Shurong Wang
- Department of Ophthalmology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yan Zhang
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
28
|
Kiyozuka K, Zhao X, Konishi A, Minamishima YA, Obinata H. Apolipoprotein M supports S1P production and conservation and mediates prolonged Akt activation via S1PR1 and S1PR3. J Biochem 2023; 174:253-266. [PMID: 37098187 DOI: 10.1093/jb/mvad037] [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: 01/21/2023] [Revised: 04/11/2023] [Accepted: 04/19/2023] [Indexed: 04/27/2023] Open
Abstract
Sphingosine 1-phosphate (S1P) is one of the lipid mediators involved in diverse physiological functions. S1P circulates in blood and lymph bound to carrier proteins. Three S1P carrier proteins have been reported, albumin, apolipoprotein M (ApoM) and apolipoprotein A4 (ApoA4). The carrier-bound S1P exerts its functions via specific S1P receptors (S1PR1-5) on target cells. Previous studies showed several differences in physiological functions between albumin-bound S1P and ApoM-bound S1P. However, molecular mechanisms underlying the carrier-dependent differences have not been clarified. In addition, ApoA4 is a recently identified S1P carrier protein, and its functional differences from albumin and ApoM have not been addressed. Here, we compared the three carrier proteins in the processes of S1P degradation, release from S1P-producing cells and receptor activation. ApoM retained S1P more stable than albumin and ApoA4 in the cell culture medium when compared in the equimolar amounts. ApoM facilitated theS1P release from endothelial cells most efficiently. Furthermore, ApoM-bound S1P showed a tendency to induce prolonged activation of Akt via S1PR1 and S1PR3. These results suggest that the carrier-dependent functional differences of S1P are partly ascribed to the differences in the S1P stability, S1P-releasing efficiency and signaling duration.
Collapse
Key Words
- Apolipoprotein A4
- Apolipoprotein M
- LC–MS/MS
- Sphingosine 1-phosphate.Abbreviations: ApoA4, Apolipoprotein A4; ApoM, Apolipoprotein M; CHO, Chinese hamster ovary; ERK, Extracellular signal-regulated kinase; LC–MS/MS, Liquid chromatography–tandem mass spectrometry; LPP, Lipid phosphate phosphatase; Mfsd2b, Multiple facilitator superfamily domain containing 2B; PBS, Phosphate-buffered saline; S1P, Sphingosine 1-phosphate; S1PR1, Sphingosine 1-phosphate receptor 1; S1PR3, Sphingosine 1-phosphate receptor 3; SphK, Sphingosine kinase; Spns2, Spinster homolog 2; TBS-T, Tris-buffed saline containing 0.1% Tween20
Collapse
Affiliation(s)
- Keisuke Kiyozuka
- Department of Biochemistry, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Xian Zhao
- Department of Biochemistry, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Akimitsu Konishi
- Department of Biochemistry, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Yoji Andrew Minamishima
- Department of Biochemistry, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Hideru Obinata
- Education and Research Support Center, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| |
Collapse
|
29
|
Tonami K, Hayashi T, Uchijima Y, Kanai M, Yura F, Mada J, Sugahara K, Kurihara Y, Kominami Y, Ushijima T, Takubo N, Liu X, Tozawa H, Kanai Y, Tokihiro T, Kurihara H. Coordinated linear and rotational movements of endothelial cells compartmentalized by VE-cadherin drive angiogenic sprouting. iScience 2023; 26:107051. [PMID: 37426350 PMCID: PMC10329149 DOI: 10.1016/j.isci.2023.107051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/22/2023] [Accepted: 06/01/2023] [Indexed: 07/11/2023] Open
Abstract
Angiogenesis is a sequential process to extend new blood vessels from preexisting ones by sprouting and branching. During angiogenesis, endothelial cells (ECs) exhibit inhomogeneous multicellular behaviors referred to as "cell mixing," in which ECs repetitively exchange their relative positions, but the underlying mechanism remains elusive. Here we identified the coordinated linear and rotational movements potentiated by cell-cell contact as drivers of sprouting angiogenesis using in vitro and in silico approaches. VE-cadherin confers the coordinated linear motility that facilitated forward sprout elongation, although it is dispensable for rotational movement, which was synchronous without VE-cadherin. Mathematical modeling recapitulated the EC motility in the two-cell state and angiogenic morphogenesis with the effects of VE-cadherin-knockout. Finally, we found that VE-cadherin-dependent EC compartmentalization potentiated branch elongations, and confirmed this by mathematical simulation. Collectively, we propose a way to understand angiogenesis, based on unique EC behavioral properties that are partially dependent on VE-cadherin function.
Collapse
Affiliation(s)
- Kazuo Tonami
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Chiyoda-ku, Tokyo 102-0076, Japan
| | - Tatsuya Hayashi
- Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Chiyoda-ku, Tokyo 102-0076, Japan
- Graduate School of Mathematical Science, The University of Tokyo, 3-8-1, Komaba, Meguro-ku, Tokyo 153-8914, Japan
- Faculty of Science and Engineering, Chuo University, 1-13-27 Kasuga, Bunkyo-ku, Tokyo 112-8551, Japan
| | - Yasunobu Uchijima
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Masahiro Kanai
- Department of Education and Creation Engineering, Kurume Institute of Technology, 2228-66 Kamitsu-machi, Kurume, Fukuoka 830-0052, Japan
| | - Fumitaka Yura
- Department of Complex and Intelligent Systems, School of Systems Information Science, Future University Hakodate, 116-2 Kamedanakano-cho, Hakodate, Hokkaido 041-8655, Japan
| | - Jun Mada
- College of Industrial Technology, Nihon University, 2-11-1 Shin-ei, Narashino, Chiba 275-8576, Japan
| | - Kei Sugahara
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yukiko Kurihara
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yuri Kominami
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1, Yayoi, Bunkyo-ku, Tokyo 113-865, Japan
| | - Toshiyuki Ushijima
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Naoko Takubo
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Chiyoda-ku, Tokyo 102-0076, Japan
- Isotope Science Center, The University of Tokyo, 2-11-16, Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Xiaoxiao Liu
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hideto Tozawa
- Department of Chemistry, Graduate School of Science, the University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yoshimitsu Kanai
- Cell Biology and Anatomy, Graduate School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama 641-8509, Japan
| | - Tetsuji Tokihiro
- Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Chiyoda-ku, Tokyo 102-0076, Japan
- Graduate School of Mathematical Science, The University of Tokyo, 3-8-1, Komaba, Meguro-ku, Tokyo 153-8914, Japan
- Department of Mathematical Engineering, Faculty of Engineering, Musashino University, 3-3-3 Ariake, Koto-ku, Tokyo 135-8181, Japan
| | - Hiroki Kurihara
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Chiyoda-ku, Tokyo 102-0076, Japan
| |
Collapse
|
30
|
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
|
31
|
Li Y, Li B, Chen WD, Wang YD. Role of G-protein coupled receptors in cardiovascular diseases. Front Cardiovasc Med 2023; 10:1130312. [PMID: 37342437 PMCID: PMC10277692 DOI: 10.3389/fcvm.2023.1130312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 05/09/2023] [Indexed: 06/22/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death globally, with CVDs accounting for nearly 30% of deaths worldwide each year. G-protein-coupled receptors (GPCRs) are the most prominent family of receptors on the cell surface, and play an essential regulating cellular physiology and pathology. Some GPCR antagonists, such as β-blockers, are standard therapy for the treatment of CVDs. In addition, nearly one-third of the drugs used to treat CVDs target GPCRs. All the evidence demonstrates the crucial role of GPCRs in CVDs. Over the past decades, studies on the structure and function of GPCRs have identified many targets for the treatment of CVDs. In this review, we summarize and discuss the role of GPCRs in the function of the cardiovascular system from both vascular and heart perspectives, then analyze the complex ways in which multiple GPCRs exert regulatory functions in vascular and heart diseases. We hope to provide new ideas for the treatment of CVDs and the development of novel drugs.
Collapse
Affiliation(s)
- Yuanqiang Li
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Boyu Li
- Department of Gastroenterology and Hematology, The People's Hospital of Hebi, Henan, China
| | - Wei-Dong Chen
- Key Laboratory of Receptors-Mediated Gene Regulation and Drug Discovery, School of Basic Medical Science, Inner Mongolia Medical University, Hohhot, China
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Medicine, The People’s Hospital of Hebi, Henan University, Kaifeng, China
| | - Yan-Dong Wang
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| |
Collapse
|
32
|
Aitken C, Mehta V, Schwartz MA, Tzima E. Mechanisms of endothelial flow sensing. NATURE CARDIOVASCULAR RESEARCH 2023; 2:517-529. [PMID: 39195881 DOI: 10.1038/s44161-023-00276-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 04/14/2023] [Indexed: 08/29/2024]
Abstract
Fluid shear stress plays a key role in sculpting blood vessels during development, in adult vascular homeostasis and in vascular pathologies. During evolution, endothelial cells evolved several mechanosensors that convert physical forces into biochemical signals, a process termed mechanotransduction. This Review discusses our understanding of endothelial flow sensing and suggests important questions for future investigation.
Collapse
Affiliation(s)
- Claire Aitken
- Wellcome Centre for Human Genetics, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Vedanta Mehta
- Wellcome Centre for Human Genetics, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Martin A Schwartz
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, and Departments of Cell Biology and Biomedical Engineering, Yale University, New Haven, CT, USA.
| | - Ellie Tzima
- Wellcome Centre for Human Genetics, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
33
|
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
|
34
|
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
|
35
|
Wang N, Li JY, Zeng B, Chen GL. Sphingosine-1-Phosphate Signaling in Cardiovascular Diseases. Biomolecules 2023; 13:biom13050818. [PMID: 37238688 DOI: 10.3390/biom13050818] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/07/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
Sphingosine-1-phosphate (S1P) is an important sphingolipid molecule involved in regulating cardiovascular functions in physiological and pathological conditions by binding and activating the three G protein-coupled receptors (S1PR1, S1PR2, and S1PR3) expressed in endothelial and smooth muscle cells, as well as cardiomyocytes and fibroblasts. It exerts its actions through various downstream signaling pathways mediating cell proliferation, migration, differentiation, and apoptosis. S1P is essential for the development of the cardiovascular system, and abnormal S1P content in the circulation is involved in the pathogenesis of cardiovascular disorders. This article reviews the effects of S1P on cardiovascular function and signaling mechanisms in different cell types in the heart and blood vessels under diseased conditions. Finally, we look forward to more clinical findings with approved S1PR modulators and the development of S1P-based therapies for cardiovascular diseases.
Collapse
Affiliation(s)
- Na Wang
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
| | - Jing-Yi Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
| | - Bo Zeng
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
| | - Gui-Lan Chen
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
36
|
Niaudet C, Jung B, Kuo A, Swendeman S, Bull E, Seno T, Crocker R, Fu Z, Smith LEH, Hla T. Therapeutic activation of endothelial sphingosine-1-phosphate receptor 1 by chaperone-bound S1P suppresses proliferative retinal neovascularization. EMBO Mol Med 2023; 15:e16645. [PMID: 36912000 PMCID: PMC10165359 DOI: 10.15252/emmm.202216645] [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: 07/25/2022] [Revised: 02/15/2023] [Accepted: 02/20/2023] [Indexed: 03/14/2023] Open
Abstract
Sphingosine-1-phosphate (S1P), the circulating HDL-bound lipid mediator that acts via S1P receptors (S1PR), is required for normal vascular development. The role of this signaling axis in vascular retinopathies is unclear. Here, we show in a mouse model of oxygen-induced retinopathy (OIR) that endothelial overexpression of S1pr1 suppresses while endothelial knockout of S1pr1 worsens neovascular tuft formation. Furthermore, neovascular tufts are increased in Apom-/- mice which lack HDL-bound S1P while they are suppressed in ApomTG mice which have more circulating HDL-S1P. These results suggest that circulating HDL-S1P activation of endothelial S1PR1 suppresses neovascular pathology in OIR. Additionally, systemic administration of ApoM-Fc-bound S1P or a small-molecule Gi-biased S1PR1 agonist suppressed neovascular tuft formation. Circulating HDL-S1P activation of endothelial S1PR1 may be a key protective mechanism to guard against neovascular retinopathies that occur not only in premature infants but also in diabetic patients and aging people.
Collapse
Affiliation(s)
- Colin Niaudet
- Department of Surgery, Vascular Biology Program, Boston Children's HospitalHarvard Medical SchoolBostonMAUSA
| | - Bongnam Jung
- Department of Surgery, Vascular Biology Program, Boston Children's HospitalHarvard Medical SchoolBostonMAUSA
| | - Andrew Kuo
- Department of Surgery, Vascular Biology Program, Boston Children's HospitalHarvard Medical SchoolBostonMAUSA
| | - Steven Swendeman
- Department of Surgery, Vascular Biology Program, Boston Children's HospitalHarvard Medical SchoolBostonMAUSA
| | - Edward Bull
- Department of Ophthalmology, Boston Children's HospitalHarvard Medical SchoolBostonMAUSA
| | - Takahiro Seno
- Department of Surgery, Vascular Biology Program, Boston Children's HospitalHarvard Medical SchoolBostonMAUSA
| | - Reed Crocker
- Department of Surgery, Vascular Biology Program, Boston Children's HospitalHarvard Medical SchoolBostonMAUSA
| | - Zhongjie Fu
- Department of Ophthalmology, Boston Children's HospitalHarvard Medical SchoolBostonMAUSA
| | - Lois E H Smith
- Department of Ophthalmology, Boston Children's HospitalHarvard Medical SchoolBostonMAUSA
| | - Timothy Hla
- Department of Surgery, Vascular Biology Program, Boston Children's HospitalHarvard Medical SchoolBostonMAUSA
| |
Collapse
|
37
|
Davies EM, Gurung R, Le KQ, Roan KT, Harvey RP, Mitchell GM, Schwarz Q, Mitchell CA. PI(4,5)P 2-dependent regulation of endothelial tip cell specification contributes to angiogenesis. SCIENCE ADVANCES 2023; 9:eadd6911. [PMID: 37000875 PMCID: PMC10065449 DOI: 10.1126/sciadv.add6911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 02/24/2023] [Indexed: 06/19/2023]
Abstract
Dynamic positioning of endothelial tip and stalk cells, via the interplay between VEGFR2 and NOTCH signaling, is essential for angiogenesis. VEGFR2 activates PI3K, which phosphorylates PI(4,5)P2 to PI(3,4,5)P3, activating AKT; however, PI3K/AKT does not direct tip cell specification. We report that PI(4,5)P2 hydrolysis by the phosphoinositide-5-phosphatase, INPP5K, contributes to angiogenesis. INPP5K ablation disrupted tip cell specification and impaired embryonic angiogenesis associated with enhanced DLL4/NOTCH signaling. INPP5K degraded a pool of PI(4,5)P2 generated by PIP5K1C phosphorylation of PI(4)P in endothelial cells. INPP5K ablation increased PI(4,5)P2, thereby releasing β-catenin from the plasma membrane, and concurrently increased PI(3,4,5)P3-dependent AKT activation, conditions that licensed DLL4/NOTCH transcription. Suppression of PI(4,5)P2 in INPP5K-siRNA cells by PIP5K1C-siRNA, restored β-catenin membrane localization and normalized AKT signaling. Pharmacological NOTCH or AKT inhibition in vivo or genetic β-catenin attenuation rescued angiogenesis defects in INPP5K-null mice. Therefore, PI(4,5)P2 is critical for β-catenin/DLL4/NOTCH signaling, which governs tip cell specification during angiogenesis.
Collapse
Affiliation(s)
- Elizabeth M. Davies
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Victoria 3800, Australia
| | - Rajendra Gurung
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Victoria 3800, Australia
| | - Kai Qin Le
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Victoria 3800, Australia
| | - Katherine T. T. Roan
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Victoria 3800, Australia
| | - Richard P. Harvey
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales 2010, Australia
- School of Clinical Medicine and School of Biotechnology and Biomolecular Science, University of New South Wales, Kensington, New South Wales 2052, Australia
| | - Geraldine M. Mitchell
- O’Brien Institute Department of St Vincent’s Institute and University of Melbourne, Department of Surgery, St. Vincent’s Hospital, Fitzroy, Victoria 3065, Australia
- Health Sciences Faculty, Australian Catholic University, Fitzroy, Victoria 3065, Australia
| | - Quenten Schwarz
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, South Australia 5001, Australia
| | - Christina A. Mitchell
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Victoria 3800, Australia
| |
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
|
Angiogenesis Model of Cornea to Understand the Role of Sphingosine 1-Phosphate. Methods Mol Biol 2023; 2625:231-240. [PMID: 36653647 DOI: 10.1007/978-1-0716-2966-6_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The bioactive sphingolipid sphingosine 1-phosphate (S1P) and its five cognate receptors (S1PR1-5) have been implicated to play important role in multiple aspects of human physiology and diseases. The S1P-S1PR1 signaling axis in endothelial cells is crucial for establishing flow competent blood vessels. The role of S1P in neovascular pathology is of great interest and is evolving as a promising target for treatment. Here we describe an easy and affordable in vivo model of corneal neovascularization using an alkali chemical burn to the cornea. This method gives a consistent and easy-to-quantitate procedure for neovascularization and angiogenesis studies.
Collapse
|
40
|
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
|
41
|
Li X, Yuan F, Zhou L. Organ Crosstalk in Acute Kidney Injury: Evidence and Mechanisms. J Clin Med 2022; 11:jcm11226637. [PMID: 36431113 PMCID: PMC9693488 DOI: 10.3390/jcm11226637] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/31/2022] [Accepted: 11/03/2022] [Indexed: 11/11/2022] Open
Abstract
Acute kidney injury (AKI) is becoming a public health problem worldwide. AKI is usually considered a complication of lung, heart, liver, gut, and brain disease, but recent findings have supported that injured kidney can also cause dysfunction of other organs, suggesting organ crosstalk existence in AKI. However, the organ crosstalk in AKI and the underlying mechanisms have not been broadly reviewed or fully investigated. In this review, we summarize recent clinical and laboratory findings of organ crosstalk in AKI and highlight the related molecular mechanisms. Moreover, their crosstalk involves inflammatory and immune responses, hemodynamic change, fluid homeostasis, hormone secretion, nerve reflex regulation, uremic toxin, and oxidative stress. Our review provides important clues for the intervention for AKI and investigates important therapeutic potential from a new perspective.
Collapse
|
42
|
Zheng Z, Lei C, Liu H, Jiang M, Zhou Z, Zhao Y, Yu CY, Wei H. A ROS-Responsive Liposomal Composite Hydrogel Integrating Improved Mitochondrial Function and Pro-Angiogenesis for Efficient Treatment of Myocardial Infarction. Adv Healthc Mater 2022; 11:e2200990. [PMID: 35848825 DOI: 10.1002/adhm.202200990] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/07/2022] [Indexed: 01/27/2023]
Abstract
Mitochondrial dysfunction of cardiomyocytes (CMs) has been identified as a significant pathogenesis of early myocardial infarction (MI). However, only a few agents or strategies have been developed to improve mitochondrial dysfunction for the effective MI treatment. Herein, a reactive oxygen species (ROS)-responsive PAMB-G-TK/4-arm-PEG-SG hydrogel is developed for localized drug-loaded liposome delivery. Notably, the liposomes contain both elamipretide (SS-31) and sphingosine-1-phosphate (S1P), where SS-31 acts as an inhibitor of mitochondrial oxidative damage and S1P as a signaling molecule for activating angiogenesis. Liposome-encapsulated PAMB-G-TK/4-arm-PEG-SG hydrogels demonstrate myocardium-like mechanical strength and electrical conductivity, and ROS-sensitive release of SS-31 and S1P-loaded liposomes. Further liposomal release of SS-31, which can target cytochrome c in the mitochondrial inner membrane of damaged CMs, inhibits pathological ROS production, improving mitochondrial dysfunction. Meanwhile, S1P released from the liposome induces endothelial cell angiogenesis by activating the S1PR1/PI3K/Akt pathway. In a rat MI model, the resulting liposomal composite hydrogel improves cardiac function by scavenging excess ROS, improving mitochondrial dysfunction, and promoting angiogenesis. This study reports for the first time a liposomal composite hydrogel that can directly target mitochondria of damaged CMs for a feedback-regulated release of encapsulated liposomes to consume the overproduced pathological ROS for improved CM activity and enhanced MI treatment.
Collapse
Affiliation(s)
- Zhi Zheng
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang, Hunan, 421001, China
| | - Cai Lei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang, Hunan, 421001, China
| | - Hongbing Liu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang, Hunan, 421001, China
| | - Mingchao Jiang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang, Hunan, 421001, China
| | - Zongtao Zhou
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang, Hunan, 421001, China
| | - Yuqi Zhao
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang, Hunan, 421001, China
| | - Cui-Yun Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang, Hunan, 421001, China
| | - Hua Wei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang, Hunan, 421001, China
| |
Collapse
|
43
|
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
|
44
|
Alshaikh RA, Ryan KB, Waeber C. Sphingosine 1-phosphate, a potential target in neovascular retinal disease. Br J Ophthalmol 2022; 106:1187-1195. [PMID: 33962970 DOI: 10.1136/bjophthalmol-2021-319115] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/17/2021] [Accepted: 04/23/2021] [Indexed: 06/12/2023]
Abstract
Neovascular ocular diseases (such as age-related macular degeneration, diabetic retinopathy and retinal vein occlusion) are characterised by common pathological processes that contribute to disease progression. These include angiogenesis, oedema, inflammation, cell death and fibrosis. Currently available therapies target the effects of vascular endothelial growth factor (VEGF), the main mediator of pathological angiogenesis. Unfortunately, VEGF blockers are expensive biological therapeutics that necessitate frequent intravitreal administration and are associated with multiple adverse effects. Thus, alternative treatment options associated with fewer side effects are required for disease management. This review introduces sphingosine 1-phosphate (S1P) as a potential pharmacological target for the treatment of neovascular ocular pathologies. S1P is a sphingolipid mediator that controls cellular growth, differentiation, survival and death. S1P actions are mediated by five G protein-coupled receptors (S1P1-5 receptors) which are abundantly expressed in all retinal and subretinal structures. The action of S1P on S1P1 receptors can reduce angiogenesis, increase endothelium integrity, reduce photoreceptor apoptosis and protect the retina against neurodegeneration. Conversely, S1P2 receptor signalling can increase neovascularisation, disrupt endothelial junctions, stimulate VEGF release, and induce retinal cell apoptosis and degeneration of neural retina. The aim of this review is to thoroughly discuss the role of S1P and its different receptor subtypes in angiogenesis, inflammation, apoptosis and fibrosis in order to determine which of these S1P-mediated processes may be targeted therapeutically.
Collapse
Affiliation(s)
- Rasha A Alshaikh
- School of Pharmacy, University College Cork, Cork, Ireland
- Department of Pharmaceutical Technology, Tanta University, Tanta, Egypt
| | - Katie B Ryan
- School of Pharmacy, University College Cork, Cork, Ireland
- SSPC The SFI Research Centre for Pharmaceuticals, School of Pharmacy, University College Cork, Cork, Ireland
| | - Christian Waeber
- School of Pharmacy, University College Cork, Cork, Ireland
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| |
Collapse
|
45
|
Le TNU, Nguyen TQ, Kalailingam P, Nguyen YTK, Sukumar VK, Tan CKH, Tukijan F, Couty L, Hasan Z, Del Gaudio I, Wenk MR, Cazenave-Gassiot A, Camerer E, Nguyen LN. Mfsd2b and Spns2 are essential for maintenance of blood vessels during development and in anaphylactic shock. Cell Rep 2022; 40:111208. [PMID: 35977478 DOI: 10.1016/j.celrep.2022.111208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 05/23/2022] [Accepted: 07/21/2022] [Indexed: 01/22/2023] Open
Abstract
Sphingosine-1-phosphate (S1P) is a potent lipid mediator that is secreted by several cell types. We recently showed that Mfsd2b is an S1P transporter from hematopoietic cells that contributes approximately 50% plasma S1P. Here we report the characterization of compound deletion of Mfsd2b and Spns2, another S1P transporter active primarily in endothelial cells. Global deletion of Mfsd2b and Spns2 (global double knockout [gDKO]) results in embryonic lethality beyond embryonic day 14.5 (E14.5), with severe hemorrhage accompanied by defects of tight junction proteins, indicating that Mfsd2b and Spns2 provide S1P for signaling, which is essential for blood vessel integrity. Compound postnatal deletion of Mfsd2b and Spns2 using Mx1Cre (ctDKO-Mx1Cre) results in maximal 80% reduction of plasma S1P. ctDKO-Mx1Cre mice exhibit severe susceptibility to anaphylaxis, indicating that S1P from Mfsd2b and Spns2 is indispensable for vascular homeostasis. Our results show that S1P export from Mfsd2b and Spns2 is essential for developing and mature vasculature.
Collapse
Affiliation(s)
- Thanh Nha Uyen Le
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| | - Toan Q Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| | - Pazhanichamy Kalailingam
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| | - Yen Thi Kim Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| | - Viresh Krishnan Sukumar
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| | - Clarissa Kai Hui Tan
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| | - Farhana Tukijan
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| | - Ludovic Couty
- Université Paris Cité, PARCC, INSERM U970, 56 Rue Leblanc, 75015 Paris, France
| | - Zafrul Hasan
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| | - Ilaria Del Gaudio
- Université Paris Cité, PARCC, INSERM U970, 56 Rue Leblanc, 75015 Paris, France
| | - Markus R Wenk
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore; Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, Singapore 117456, Singapore
| | - Amaury Cazenave-Gassiot
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore; Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, Singapore 117456, Singapore
| | - Eric Camerer
- Université Paris Cité, PARCC, INSERM U970, 56 Rue Leblanc, 75015 Paris, France
| | - Long N Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore; Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, Singapore 117456, Singapore; Cardiovascular Disease Research (CVD) Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore; Immunology Program, Life Sciences Institute, 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
|
Abstract
COVID-19 is a primary respiratory illness that is frequently complicated by systemic involvement of the vasculature. Vascular involvement leads to an array of complications ranging from thrombosis to pulmonary edema secondary to loss of barrier function. This review will address the vasculopathy of COVID-19 with a focus on the role of the endothelium in orchestrating the systemic response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. The endothelial receptor systems and molecular pathways activated in the setting of COVID-19 and the consequences of these inflammatory and prothrombotic changes on endothelial cell function will be discussed. The sequelae of COVID-19 vascular involvement at the level of organ systems will also be addressed, with an emphasis on the pulmonary vasculature but with consideration of effects on other vascular beds. The dramatic changes in endothelial phenotypes associated with COVID-19 has enabled the identification of biomarkers that could help guide therapy and predict outcomes. Knowledge of vascular pathogenesis in COVID-19 has also informed therapeutic approaches that may control its systemic sequelae. Because our understanding of vascular response in COVID-19 continues to evolve, we will consider areas of controversy, such as the extent to which SARS-CoV-2 directly infects endothelium and the degree to which vascular responses to SARS-CoV-2 are unique or common to those of other viruses capable of causing severe respiratory disease. This conceptual framework describing how SARS-CoV-2 infection affects endothelial inflammation, prothrombotic transformation, and barrier dysfunction will provide a context for interpreting new information as it arises addressing the vascular complications of COVID-19.
Collapse
Affiliation(s)
| | | | - Alec A Schmaier
- Division of Hemostasis and Thrombosis and
- Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| |
Collapse
|
47
|
Yazbeck P, Cullere X, Bennett P, Yajnik V, Wang H, Kawada K, Davis V, Parikh A, Kuo A, Mysore V, Hla T, Milstone D, Mayadas TN. DOCK4 Regulation of Rho GTPases Mediates Pulmonary Vascular Barrier Function. Arterioscler Thromb Vasc Biol 2022; 42:886-902. [PMID: 35477279 PMCID: PMC9233130 DOI: 10.1161/atvbaha.122.317565] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 04/12/2022] [Indexed: 01/10/2023]
Abstract
BACKGROUND The vascular endothelium maintains tissue-fluid homeostasis by controlling the passage of large molecules and fluid between the blood and interstitial space. The interaction of catenins and the actin cytoskeleton with VE-cadherin (vascular endothelial cadherin) is the primary mechanism for stabilizing AJs (adherens junctions), thereby preventing lung vascular barrier disruption. Members of the Rho (Ras homology) family of GTPases and conventional GEFs (guanine exchange factors) of these GTPases have been demonstrated to play important roles in regulating endothelial permeability. Here, we evaluated the role of DOCK4 (dedicator of cytokinesis 4)-an unconventional Rho family GTPase GEF in vascular function. METHODS We generated mice deficient in DOCK4' used DOCK4 silencing and reconstitution approaches in human pulmonary artery endothelial cells' used assays to evaluate protein localization, endothelial cell permeability, and small GTPase activation. RESULTS Our data show that DOCK4-deficient mice are viable. However, these mice have hemorrhage selectively in the lung, incomplete smooth muscle cell coverage in pulmonary vessels, increased basal microvascular permeability, and impaired response to S1P (sphingosine-1-phosphate)-induced reversal of thrombin-induced permeability. Consistent with this, DOCK4 rapidly translocates to the cell periphery and associates with the detergent-insoluble fraction following S1P treatment, and its absence prevents S1P-induced Rac-1 activation and enhancement of barrier function. Moreover, DOCK4-silenced pulmonary artery endothelial cells exhibit enhanced basal permeability in vitro that is associated with enhanced Rho GTPase activation. CONCLUSIONS Our findings indicate that DOCK4 maintains AJs necessary for lung vascular barrier function by establishing the normal balance between RhoA (Ras homolog family member A) and Rac-1-mediated actin cytoskeleton remodeling, a previously unappreciated function for the atypical GEF family of molecules. Our studies also identify S1P as a potential upstream regulator of DOCK4 activity.
Collapse
Affiliation(s)
- Pascal Yazbeck
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| | - Xavier Cullere
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| | - Paul Bennett
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| | - Vijay Yajnik
- Department of Medicine, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02445
| | - Huan Wang
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| | - Kenji Kawada
- Department of Medicine, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02445
| | - Vanessa Davis
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| | - Asit Parikh
- Department of Medicine, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02445
| | - Andrew Kuo
- Vascular Biology Program, Boston Children’s Hospital and Harvard Medical School, Boston, MA 20115
| | - Vijayashree Mysore
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| | - Timothy Hla
- Vascular Biology Program, Boston Children’s Hospital and Harvard Medical School, Boston, MA 20115
| | - David Milstone
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| | - Tanya N. Mayadas
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| |
Collapse
|
48
|
Wang F, Zhang X, Liu Y, Li Z, Wei R, Zhang Y, Zhang R, Khan S, Yong VW, Xue M. Neuroprotection by Ozanimod Following Intracerebral Hemorrhage in Mice. Front Mol Neurosci 2022; 15:927150. [PMID: 35782389 PMCID: PMC9242004 DOI: 10.3389/fnmol.2022.927150] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 05/23/2022] [Indexed: 12/26/2022] Open
Abstract
The destruction of the blood-brain barrier (BBB) after intracerebral hemorrhage (ICH) is associated with poor prognosis. Modulation of sphingosine 1-phosphate receptor (S1PR) may improve outcomes from ICH. Ozanimod (RPC-1063) is a newly developed S1PR regulator which can selectively modulate type 1/5 sphingosine receptors. Here, we studied the impact of Ozanimod on neuroprotection in an experimental mouse model of ICH, induced by injecting collagenase type VII into the basal ganglia. Ozanimod was administered by gavage 2 h after surgery and once a day thereafter until sacrifice. The results demonstrate that Ozanimod treatment improved neurobehavioral deficits in mice and decreased weight loss after ICH. Ozanimod significantly reduced the density of activated microglia and infiltrated neutrophils in the perihematoma region. Furthermore, Ozanimod reduced hematoma volume and water content of the ICH brain. The results of TUNEL staining indicate that Ozanimod mitigated brain cell death. The quantitative data of Evans blue (EB) staining showed that Ozanimod reduced EB dye leakage. Overall, Ozanimod reduces the destruction of the BBB and exert neuroprotective roles following ICH in mice.
Collapse
Affiliation(s)
- Fei Wang
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, China
| | - Xiangyu Zhang
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, China
| | - Yang Liu
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, China
| | - Zhe Li
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, China
| | - Ruixue Wei
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, China
| | - Yan Zhang
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, China
| | - Ruiyi Zhang
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, China
| | - Suliman Khan
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, China
| | - V. Wee Yong
- Department of Clinical Neurosciences, The Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- *Correspondence: V. Wee Yong,
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, China
- Mengzhou Xue,
| |
Collapse
|
49
|
Spampinato SF, Sortino MA, Salomone S. Sphingosine-1-phosphate and Sphingosine-1-phosphate receptors in the cardiovascular system: pharmacology and clinical implications. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2022; 94:95-139. [PMID: 35659378 DOI: 10.1016/bs.apha.2022.02.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Sphingosine-1-phosphate (S1P) is a lipid that binds and activates five distinct receptor subtypes, S1P1, S1P2, S1P3, S1P4, S1P5, widely expressed in different cells, tissues and organs. In the cardiovascular system these receptors have been extensively studied, but no drug acting on them has been approved so far for treating cardiovascular diseases. In contrast, a number of S1P receptor agonists are approved as immunomodulators, mainly for multiple sclerosis, because of their action on lymphocyte trafficking. This chapter summarizes the available information on S1P receptors in the cardiovascular system and discusses their potential for treating cardiovascular conditions and/or their role on the clinical pharmacology of drugs so far approved for non-cardiovascular conditions. Basic research has recently produced data useful to understand the molecular pharmacology of S1P and S1P receptors, regarding biased agonism, S1P storage, release and vehiculation and chaperoning by lipoproteins, paracrine actions, intracellular non-receptorial S1P actions. On the other hand, the approval of fingolimod and newer generation S1P receptor ligands as immunomodulators, provides information on a number of clinical observations on the impact of these drugs on cardiovascular system which need to be integrated with preclinical data. S1P receptors are potential targets for prevention and treatment of major cardiovascular conditions, including hypertension, myocardial infarction, heart failure and stroke.
Collapse
Affiliation(s)
| | - Maria Angela Sortino
- Department of Biomedical and Biotechnological Science, University of Catania, Catania, Italy
| | - Salvatore Salomone
- Department of Biomedical and Biotechnological Science, University of Catania, Catania, Italy.
| |
Collapse
|
50
|
Sphingosine 1-phosphate receptor-targeted therapeutics in rheumatic diseases. Nat Rev Rheumatol 2022; 18:335-351. [PMID: 35508810 DOI: 10.1038/s41584-022-00784-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2022] [Indexed: 02/07/2023]
Abstract
Sphingosine 1-phosphate (S1P), which acts via G protein-coupled S1P receptors (S1PRs), is a bioactive lipid essential for vascular integrity and lymphocyte trafficking. The S1P-S1PR signalling axis is a key component of the inflammatory response in autoimmune rheumatic diseases. Several drugs that target S1PRs have been approved for the treatment of multiple sclerosis and inflammatory bowel disease and are under clinical testing for patients with systemic lupus erythematosus (SLE). Preclinical studies support the hypothesis that targeting the S1P-S1PR axis would be beneficial to patients with SLE, rheumatoid arthritis (RA) and systemic sclerosis (SSc) by reducing pathological inflammation. Whereas most preclinical research and development efforts are focused on reducing lymphocyte trafficking, protective effects of circulating S1P on endothelial S1PRs, which maintain the vascular barrier and enable blood circulation while dampening leukocyte extravasation, have been largely overlooked. In this Review, we take a holistic view of S1P-S1PR signalling in lymphocyte and vascular pathobiology. We focus on the potential of S1PR modulators for the treatment of SLE, RA and SSc and summarize the rationale, pathobiology and evidence from preclinical models and clinical studies. Improved understanding of S1P pathobiology in autoimmune rheumatic diseases and S1PR therapeutic modulation is anticipated to lead to efficacious and safer management of these diseases.
Collapse
|