1
|
Deng H, Eichmann A, Schwartz MA. Fluid Shear Stress-Regulated Vascular Remodeling: Past, Present, and Future. Arterioscler Thromb Vasc Biol 2025; 45:882-900. [PMID: 40207366 DOI: 10.1161/atvbaha.125.322557] [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: 04/11/2025]
Abstract
The vascular system remodels throughout life to ensure adequate perfusion of tissues as they grow, regress, or change metabolic activity. Angiogenesis, the sprouting of new blood vessels to expand the capillary network, versus regression, in which endothelial cells die or migrate away to remove unneeded capillaries, controls capillary density. In addition, upstream arteries adjust their diameters to optimize blood flow to downstream vascular beds, which is controlled primarily by vascular endothelial cells sensing fluid shear stress (FSS) from blood flow. Changes in capillary density and small artery tone lead to changes in the resistance of the vascular bed, which leads to changes in flow through the arteries that feed these small vessels. The resultant decreases or increases in FSS through these vessels then stimulate their inward or outward remodeling, respectively. This review summarizes our knowledge of endothelial FSS-dependent vascular remodeling, offering insights into potential therapeutic interventions. We first provide a historical overview, then discuss the concept of set point and mechanisms of low-FSS-mediated and high-FSS-mediated inward and outward remodeling. We then cover in vivo animal models, molecular mechanisms, and clinical implications. Understanding the mechanisms underlying physiological endothelial FSS-mediated vascular remodeling and their failure due to mutations or chronic inflammatory and metabolic stresses may lead to new therapeutic strategies to prevent or treat vascular diseases.
Collapse
Affiliation(s)
- Hanqiang Deng
- Yale Cardiovascular Research Center CT (H.D., A.E., M.A.S.), Yale University School of Medicine, New Haven, CT
- Section of Cardiovascular Medicine, Department of Internal Medicine (H.D., A.E., M.A.S.), Yale University School of Medicine, New Haven, CT
| | - Anne Eichmann
- Yale Cardiovascular Research Center CT (H.D., A.E., M.A.S.), Yale University School of Medicine, New Haven, CT
- Section of Cardiovascular Medicine, Department of Internal Medicine (H.D., A.E., M.A.S.), Yale University School of Medicine, New Haven, CT
| | - Martin A Schwartz
- Yale Cardiovascular Research Center CT (H.D., A.E., M.A.S.), Yale University School of Medicine, New Haven, CT
- Section of Cardiovascular Medicine, Department of Internal Medicine (H.D., A.E., M.A.S.), Yale University School of Medicine, New Haven, CT
- Department of Cell Biology, Yale School of Medicine, New Haven, CT (M.A.S.)
- Department of Biomedical Engineering, Yale School of Engineering, New Haven, CT (M.A.S.)
| |
Collapse
|
2
|
Mishra S, Ipe U, Nottebaum AF, Peters KG, Vestweber D. Inhibition of VE-PTP rejuvenates Schlemm's canal in aged mice and acts via Tie2. PLoS One 2025; 20:e0323615. [PMID: 40378380 DOI: 10.1371/journal.pone.0323615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 04/10/2025] [Indexed: 05/18/2025] Open
Abstract
PURPOSE Glaucoma is the leading cause of irreversible blindness worldwide and is associated with high intraocular pressure (IOP). Schlemm's canal (SC), a hybrid vessel present in the anterior part of the eye, is known to control IOP by draining aqueous humor into the systemic circulation. Formation and function of SC is supported by the tyrosine kinase receptor Tie2. Likewise, inhibition of the vascular endothelial protein tyrosine phosphatase (VE-PTP), which associates with Tie2 has similar effects. However, VE-PTP also targets VE-cadherin and several other substrates. Here, we analyzed whether Tie2 is indeed the major substrate which is responsible for the role of VE-PTP in SC function. In addition, we analyzed the function of VE-PTP in SC of the aged eye in mice. METHODOLOGY We tested the effects of the VE-PTP inhibitor AKB9778 and of VE-PTP gene inactivation on SC area and IOP in WT and in Tie2iLEC/SC-KO and VE-cadherin-Y685F mutant mice. RESULTS Pharmacologic inhibition of VE-PTP with AKB9778 increased SC area only in mice expressing Tie2. The VE-cadherin-Y685F mutation had neither an effect on SC area nor on the effects of AKB9778 on SC formation. Induced VE-PTP gene inactivation in adult mice had similar effects as AKB9778. Furthermore, we could show that AKB9778 improved SC function in aged mice as judged by increasing SC area and lowering of IOP. CONCLUSION Interference with VE-PTP function improves SC function in a strictly Tie2 dependent way and pharmacologic inhibition of VE-PTP with AKB9778 is a promising approach for improving SC function in the aged eye.
Collapse
Affiliation(s)
- Sarthak Mishra
- Department of Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Ute Ipe
- Department of Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Astrid F Nottebaum
- Department of Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Kevin G Peters
- Aerpio Therapeutics, Inc., Blue Ash, Ohio, United States of America
| | - Dietmar Vestweber
- Department of Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| |
Collapse
|
3
|
Gucciardo F, Lebeau A, Pirson S, Buntinx F, Ivanova E, Blacher S, Brouillard P, Deroye J, Baudin L, Pirnay A, Morfoisse F, Villette C, Nizet C, Lallemand F, Munaut C, Alitalo K, Geris L, Vikkula M, Gautier-Isola M, Noel A. Targeting uPARAP Modifies Lymphatic Vessel Architecture and Attenuates Lymphedema. Circulation 2025; 151:1412-1429. [PMID: 40035133 PMCID: PMC12063686 DOI: 10.1161/circulationaha.124.072093] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 02/04/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND Lymphedema is an incurable disease associated with lymphatic dysfunction that causes tissue swelling and fibrosis. We investigated whether lymphedema could be attenuated by interfering with uPARAP (urokinase plasminogen activator receptor-associated protein; Mrc2 gene), an endocytic receptor involved in fibrosis and lymphangiogenesis. METHODS We generated mice with lymphatic endothelial cell (LEC)-specific uparap deficiency and compared them with constitutive knockout mice by applying a preclinical model of secondary lymphedema (SL). Computerized methods were applied for 2-dimensional and 3-dimensional image quantifications. Cellular effects of uPARAP deletion on lymphatic permeability were assessed by small interfering RNA-mediated silencing in human dermal LECs and a pharmacologic treatment targeting ROCK (Rho-associated coiled coil containing kinase), an established regulator of cell junctions. The uPARAP and vascular endothelial cadherin partnership was investigated through proximity ligation assay, coimmunoprecipitation, and immunostaining. An in silico model was generated to analyze the fluid-absorbing function of the lymphatic vasculature. To interfere with uPARAP, its downregulation was achieved in vivo through a gapmer approach. RESULTS uparap deficiency mitigated several key pathologic features of SL, including hindlimb swelling, epidermal thickening, and the accumulation and size of adipocytes. In both global and LEC-conditional uparap-deficient mice, induction of SL led to a distinctive labyrinthine vasculature, defined herein by twisted and hyperbranched vessels with overlapping cells. This topology, mainly composed of pre-collecting vessels, correlated with reduced SL, but not with change in fibrosis, highlighting the importance of uPARAP in regulating LEC functions in a lymphedematous context. In vitro, uPARAP knockdown in LECs impaired vascular endothelial growth factor C-mediated endosomal trafficking of vascular endothelial cadherin and induced overlapping cell junctions. The pharmacologic inhibition of ROCK recapitulated cell superimposition in vitro and the labyrinthine vasculature in vivo with attenuated SL. Computational modeling of labyrinthine lymphatic vasculature supported the observation on their improved fluid-absorbing function in comparison with a normal hierarchic network. These data provide proof of concept of inducing a labyrinthine topology to treat SL. For therapeutic purposes, we validated the use of an anti-uPARAP gapmer to induce a labyrinthine vasculature and attenuate SL formation. CONCLUSIONS Our findings provide evidence that downregulating uPARAP expression can induce a beneficial remodeling of lymphatic vasculature that attenuates lymphedema through a cell junction-based mechanism, offering a novel therapeutic pathway for lymphedema.
Collapse
Affiliation(s)
- Fabrice Gucciardo
- From the Laboratory of Tumor and Development Biology, GIGA (F.G., A.L., S.P., F.B., E.I., S.B., J.D., L.B., A.P., C.M., M.G.-I., A.N.), University of Liège, Sart-Tilman, Belgium
| | - Alizée Lebeau
- From the Laboratory of Tumor and Development Biology, GIGA (F.G., A.L., S.P., F.B., E.I., S.B., J.D., L.B., A.P., C.M., M.G.-I., A.N.), University of Liège, Sart-Tilman, Belgium
| | - Sébastien Pirson
- From the Laboratory of Tumor and Development Biology, GIGA (F.G., A.L., S.P., F.B., E.I., S.B., J.D., L.B., A.P., C.M., M.G.-I., A.N.), University of Liège, Sart-Tilman, Belgium
| | - Florence Buntinx
- From the Laboratory of Tumor and Development Biology, GIGA (F.G., A.L., S.P., F.B., E.I., S.B., J.D., L.B., A.P., C.M., M.G.-I., A.N.), University of Liège, Sart-Tilman, Belgium
| | - Elitsa Ivanova
- From the Laboratory of Tumor and Development Biology, GIGA (F.G., A.L., S.P., F.B., E.I., S.B., J.D., L.B., A.P., C.M., M.G.-I., A.N.), University of Liège, Sart-Tilman, Belgium
| | - Silvia Blacher
- From the Laboratory of Tumor and Development Biology, GIGA (F.G., A.L., S.P., F.B., E.I., S.B., J.D., L.B., A.P., C.M., M.G.-I., A.N.), University of Liège, Sart-Tilman, Belgium
| | - Pascal Brouillard
- Human Molecular Genetics, de Duve Institute, University of Louvain, Brussels, Belgium (P.B., M.V.)
| | - Jonathan Deroye
- From the Laboratory of Tumor and Development Biology, GIGA (F.G., A.L., S.P., F.B., E.I., S.B., J.D., L.B., A.P., C.M., M.G.-I., A.N.), University of Liège, Sart-Tilman, Belgium
| | - Louis Baudin
- From the Laboratory of Tumor and Development Biology, GIGA (F.G., A.L., S.P., F.B., E.I., S.B., J.D., L.B., A.P., C.M., M.G.-I., A.N.), University of Liège, Sart-Tilman, Belgium
| | - Alexandra Pirnay
- From the Laboratory of Tumor and Development Biology, GIGA (F.G., A.L., S.P., F.B., E.I., S.B., J.D., L.B., A.P., C.M., M.G.-I., A.N.), University of Liège, Sart-Tilman, Belgium
| | - Florent Morfoisse
- U1297-Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, France (F.M.)
| | - Claire Villette
- Biomechanics Research Unit Department, Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, Belgium (C.V., L.G.)
| | - Christophe Nizet
- Departments of Plastic and Reconstructive Surgery (C.N.), University of Liège, Sart-Tilman, Belgium
| | | | - Carine Munaut
- From the Laboratory of Tumor and Development Biology, GIGA (F.G., A.L., S.P., F.B., E.I., S.B., J.D., L.B., A.P., C.M., M.G.-I., A.N.), University of Liège, Sart-Tilman, Belgium
| | - Kari Alitalo
- Wihuri Research Institute and Translational Cancer Medicine Program, Biomedicum, University of Helsinki, Finland (K.A.)
| | - Liesbet Geris
- Biomechanics Research Unit Department, Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, Belgium (C.V., L.G.)
| | - Miikka Vikkula
- Human Molecular Genetics, de Duve Institute, University of Louvain, Brussels, Belgium (P.B., M.V.)
- WELBIO Department, WEL Research Institute, Wavre, Belgium (M.V., A.N.)
| | - Marine Gautier-Isola
- From the Laboratory of Tumor and Development Biology, GIGA (F.G., A.L., S.P., F.B., E.I., S.B., J.D., L.B., A.P., C.M., M.G.-I., A.N.), University of Liège, Sart-Tilman, Belgium
| | - Agnès Noel
- From the Laboratory of Tumor and Development Biology, GIGA (F.G., A.L., S.P., F.B., E.I., S.B., J.D., L.B., A.P., C.M., M.G.-I., A.N.), University of Liège, Sart-Tilman, Belgium
- WELBIO Department, WEL Research Institute, Wavre, Belgium (M.V., A.N.)
| |
Collapse
|
4
|
Jiang W, Guo Z, Wang Q, Chen Z, Dong W, Liang Q, Hao Y, Pan H, Zeng C, Liu H, Wang Y. Enhanced nanoparticle delivery across vascular basement membranes of tumours using nitric oxide. Nat Biomed Eng 2025:10.1038/s41551-025-01385-w. [PMID: 40316687 DOI: 10.1038/s41551-025-01385-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 04/03/2025] [Indexed: 05/04/2025]
Abstract
The delivery of nanoparticles (NPs) into solid tumours is challenged by the tumour vascular basement membrane (BM), a critical barrier beneath the endothelium with robust mechanical properties resistant to conventional treatments. Here we propose an approach that uses nitric oxide (NO) to induce the opening of endothelial junctions, creating gaps between endothelial cells and enabling the navigation of NPs through these gaps. Subsequently, NO orchestrates a transient degradation of the BM encasing NP pools in a precise, localized action, allowing the enhanced passage of NPs into the tumour interstitial space through explosive eruptions. We have engineered a NO nanogenerator tailored for near-infrared laser-triggered on-demand NO release at tumour sites. Through breaching the BM barrier, this system results in an increase of clinical nanomedicines within the tumour, boosting the tumour suppression efficacy in both mouse and rabbit models. This approach delicately manages BM degradation, avoiding excessive degradation that might facilitate cancer metastasis. Our NO nanogenerator serves as a precise spatial catalytic degradation strategy for breaching the tumour vascular BM barrier, holding promise for NP delivery into non-tumour diseases.
Collapse
Affiliation(s)
- Wei Jiang
- National Key Laboratory of Immune Response and Immunotherapy, Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China
| | - Zixuan Guo
- National Key Laboratory of Immune Response and Immunotherapy, Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Qin Wang
- National Key Laboratory of Immune Response and Immunotherapy, Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Macao Institute of Materials Science and Engineering, Macau University of Science and Technology, Macau, China.
| | - Ziqi Chen
- National Key Laboratory of Immune Response and Immunotherapy, Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Wang Dong
- National Key Laboratory of Immune Response and Immunotherapy, Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Qirui Liang
- National Key Laboratory of Immune Response and Immunotherapy, Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China
| | - Yinghong Hao
- National Key Laboratory of Immune Response and Immunotherapy, Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Huimin Pan
- National Key Laboratory of Immune Response and Immunotherapy, Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Cici Zeng
- National Key Laboratory of Immune Response and Immunotherapy, Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Hang Liu
- National Key Laboratory of Immune Response and Immunotherapy, Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Anhui Province Engineering Research Center of Critical Electronic Materials, School of Chemistry and Chemical Engineering, Anhui University, Hefei, China.
| | - Yucai Wang
- National Key Laboratory of Immune Response and Immunotherapy, Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China.
- School of Biomedical Engineering, Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, China.
| |
Collapse
|
5
|
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
|
6
|
Vargas-Robles H, Hernández-Almaraz KB, Schnoor M. Analysis of Vascular Permeability by a Modified Miles Assay. Bio Protoc 2025; 15:e5264. [PMID: 40224658 PMCID: PMC11986696 DOI: 10.21769/bioprotoc.5264] [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: 12/19/2024] [Revised: 03/03/2025] [Accepted: 03/04/2025] [Indexed: 04/15/2025] Open
Abstract
The endothelial barrier is a semipermeable cell layer covering the inside of blood vessels that regulates the flux of ions, macromolecules, and plasma from blood to tissues. Inflammation promotes an increase in vascular permeability, which can contribute to disease if not controlled properly. Thus, it is important to understand in detail the molecular mechanisms underlying inflammatory vascular hyperpermeability. While endothelial permeability can be measured in vitro, these assays do not recapitulate precisely the in vivo vasculature. Thus, in vivo assays are required to understand the full picture of vascular permeability regulation. Here, we describe an established assay that involves injection of Evans blue dye followed by intradermal injection of agents inducing vascular permeability. This assay is relatively easy to perform and provides reliable data on permeability regulation in vivo. Key features • Step-by-step protocol to study vascular permeability in the mouse skin. • Injection of Evans blue dye followed by intradermal injection of permeability-inducing agents allows reproducible analysis of regulatory mechanisms. • This protocol allows the analysis of different substances in the same animal. • Possibility of different dye administration routes that can be compared.
Collapse
Affiliation(s)
- Hilda Vargas-Robles
- Department for Molecular Biomedicine, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), Mexico-City, Mexico
| | - Karina B. Hernández-Almaraz
- Department for Molecular Biomedicine, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), Mexico-City, Mexico
| | - Michael Schnoor
- Department for Molecular Biomedicine, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), Mexico-City, Mexico
| |
Collapse
|
7
|
Brash JT, Diez-Pinel G, Rinaldi L, Castellan RFP, Fantin A, Ruhrberg C. Endothelial transcriptomic, epigenomic and proteomic data challenge the proposed role for TSAd in vascular permeability. Angiogenesis 2025; 28:21. [PMID: 40080216 PMCID: PMC11906500 DOI: 10.1007/s10456-025-09971-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 02/23/2025] [Indexed: 03/15/2025]
Abstract
The vascular endothelial growth factor VEGF drives excessive vascular permeability to cause tissue-damaging oedema in neovascular and inflammatory diseases across multiple organs. Several molecular pathways have been implicated in VEGF-induced hyperpermeability, including binding of the VEGF-activated tyrosine kinase receptor VEGFR2 by the T-cell specific adaptor (TSAd) to recruit a SRC family kinase to induce junction opening between vascular endothelial cells (ECs). Inconsistent with a universal role for TSAd in permeability signalling, immunostaining approaches previously reported TSAd only in dermal and kidney vasculature. To address this discrepancy, we have mined publicly available omics data for expression of TSAd and other permeability-relevant signal transducers in multiple organs affected by VEGF-induced vascular permeability. Unexpectedly, TSAd transcripts were largely absent from EC single cell RNAseq data, whereas transcripts for other permeability-relevant signal transducers were detected readily. TSAd transcripts were also lacking from half of the EC bulk RNAseq datasets examined, and in the remaining datasets appeared at low levels concordant with models of leaky transcription. Epigenomic EC data located the TSAd promoter to closed chromatin in ECs, and mass spectrometry-derived EC proteomes typically lacked TSAd. By suggesting that TSAd is not actively expressed in ECs, our findings imply that TSAd is likely not critical for linking VEGFR2 to downstream signal transducers for EC junction opening.
Collapse
Affiliation(s)
- James T Brash
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK
| | - Guillermo Diez-Pinel
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK
| | - Luca Rinaldi
- Department of Biosciences, University of Milan, Via G. Celoria 26, 20133, Milan, Italy
| | - Raphael F P Castellan
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK
| | - Alessandro Fantin
- Department of Biosciences, University of Milan, Via G. Celoria 26, 20133, Milan, Italy.
| | - Christiana Ruhrberg
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK.
| |
Collapse
|
8
|
Santos F, Sum H, Yan DCL, Brewer AC. Metaboloepigenetics: Role in the Regulation of Flow-Mediated Endothelial (Dys)Function and Atherosclerosis. Cells 2025; 14:378. [PMID: 40072106 PMCID: PMC11898952 DOI: 10.3390/cells14050378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 02/26/2025] [Accepted: 03/03/2025] [Indexed: 03/15/2025] Open
Abstract
Endothelial dysfunction is the main initiating factor in atherosclerosis. Through mechanotransduction, shear stress regulates endothelial cell function in both homeostatic and diseased states. Accumulating evidence reveals that epigenetic changes play critical roles in the etiology of cardiovascular diseases, including atherosclerosis. The metabolic regulation of epigenetics has emerged as an important factor in the control of gene expression in diseased states, but to the best of our knowledge, this connection remains largely unexplored in endothelial dysfunction and atherosclerosis. In this review, we (1) summarize how shear stress (or flow) regulates endothelial (dys)function; (2) explore the epigenetic alterations that occur in the endothelium in response to disturbed flow; (3) review endothelial cell metabolism under different shear stress conditions; and (4) suggest mechanisms which may link this altered metabolism to the regulation of the endothelial epigenome by modulations in metabolite availability. We believe that metabolic regulation plays an important role in endothelial epigenetic reprogramming and could pave the way for novel metabolism-based therapeutic strategies.
Collapse
Affiliation(s)
- Francisco Santos
- School of Cardiovascular and Metabolic Medicine & Sciences, British Heart Foundation Centre of Research Excellence, Faculty of Life Sciences & Medicine, King’s College London, London SE5 9NU, UK; (F.S.); (H.S.)
| | - Hashum Sum
- School of Cardiovascular and Metabolic Medicine & Sciences, British Heart Foundation Centre of Research Excellence, Faculty of Life Sciences & Medicine, King’s College London, London SE5 9NU, UK; (F.S.); (H.S.)
| | | | - Alison C. Brewer
- School of Cardiovascular and Metabolic Medicine & Sciences, British Heart Foundation Centre of Research Excellence, Faculty of Life Sciences & Medicine, King’s College London, London SE5 9NU, UK; (F.S.); (H.S.)
| |
Collapse
|
9
|
Soehnlein O, Lutgens E, Döring Y. Distinct inflammatory pathways shape atherosclerosis in different vascular beds. Eur Heart J 2025:ehaf054. [PMID: 40036569 DOI: 10.1093/eurheartj/ehaf054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 11/01/2024] [Accepted: 01/23/2025] [Indexed: 03/06/2025] Open
Abstract
Studies suggest varying atherosclerotic cardiovascular disease (ASCVD) prevalence across arterial beds. Factors such as smoking expedite ASCVD progression in the abdominal aorta, while diabetes accelerates plaque development in lower limb arteries, and hypertension plays a significant role in ASCVD development in the coronary and carotid arteries. Moreover, superficial femoral atherosclerosis advances slower compared with atherosclerosis in coronary and carotid arteries. Furthermore, femoral atherosclerosis exhibits higher levels of ossification and calcification, but lower cholesterol concentrations compared with atherosclerotic lesions of other vascular beds. Such disparities exemplify the diverse progression of ASCVD across arterial beds, pointing towards differential mechanistic pathways in each vascular bed. Hence, this review summarizes current literature on immune-inflammatory mechanisms in various arterial beds in ASCVD to advance our understanding of this disease in an aging society with increased need of vascular bed and patient-specific treatment options.
Collapse
Affiliation(s)
- Oliver Soehnlein
- Institute of Experimental Pathology (ExPat), Center of Molecular Biology of Inflammation (ZMBE), Von-Esmarch-Str. 56, University of Münster, 48149 Münster, Germany
| | - Esther Lutgens
- Cardiovascular Medicine and Immunology, Experimental Cardiovascular Immunology Laboratory, Mayo Clinic, Rochester, MN, USA
- German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung, DZHK), Munich Heart Alliance Partner Site, Munich, Germany
| | - Yvonne Döring
- German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung, DZHK), Munich Heart Alliance Partner Site, Munich, Germany
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), Bern University Hospital, University of Bern, Bern, Switzerland
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian University, Munich, Germany
| |
Collapse
|
10
|
Kizhatil K, Clark GM, Sunderland DK, Bhandari A, Horbal LJ, Balasubramanian R, John SWM. FYN regulates aqueous humor outflow and IOP through the phosphorylation of VE-CADHERIN. Nat Commun 2025; 16:51. [PMID: 39746990 PMCID: PMC11696269 DOI: 10.1038/s41467-024-55232-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 12/03/2024] [Indexed: 01/04/2025] Open
Abstract
Schlemm's canal endothelial cells (SECs) serve as the final barrier to aqueous humor (AQH) drainage from the eye. SECs adjust permeability to AQH outflow to modulate intraocular pressure (IOP). The broad identification of IOP-related genes implicates SECs in glaucoma. However, the molecular mechanisms by which SECs sense and respond to pressure changes to regulate fluid permeability and IOP remain largely undefined. We hypothesize that mechano-responsive phosphorylation of the cell adhesion molecule VE-CADHERIN (CDH5) in SECs, by FYN and possibly other SRC family kinases, regulates adherens junction (AJ) permeability to AQH in response to IOP. On experimentally raising IOP in mouse eyes, AJ permeability, CDH5 phosphorylation, and FYN activation at the AJ all increase. FYN null mutant mice display disrupted IOP regulation and reduced AQH outflow. These findings demonstrate an important role of mechanotransducive signaling within SECs in maintaining IOP homeostasis and implicate FYN as a potential target for developing IOP-lowering treatments.
Collapse
Affiliation(s)
- Krishnakumar Kizhatil
- Department of Ophthalmology and Visual Sciences, The Ohio State Medical Center, Columbus, Ohio, 43210, USA.
| | | | | | - Aakriti Bhandari
- The Jackson Laboratory, Bar Harbor, Maine, 04609, USA
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY, USA
| | - Logan J Horbal
- The Jackson Laboratory, Bar Harbor, Maine, 04609, USA
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Simon W M John
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY, USA.
- Zuckerman Mind Brain Behavior Institute, New York, NY, 10027, USA.
| |
Collapse
|
11
|
Pal S, Su Y, Nwadozi E, Claesson-Welsh L, Richards M. Neuropilin-1 controls vascular permeability through juxtacrine regulation of endothelial adherens junctions. Angiogenesis 2024; 28:7. [PMID: 39668325 PMCID: PMC11638295 DOI: 10.1007/s10456-024-09963-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 12/01/2024] [Indexed: 12/14/2024]
Abstract
Neuropilin-1 (NRP1) regulates endothelial cell (EC) biology through modulation of vascular endothelial growth factor receptor 2 (VEGFR2) signalling by presenting VEGFA to VEGFR2. How NRP1 impacts VEGFA-mediated vascular hyperpermeability has however remained unresolved, described as exerting either a positive or a passive function. Using EC-specific Nrp1 knock-out mice, we discover that EC-expressed NRP1 exerts an organotypic role. In the ear skin, VEGFA/VEGFR2-mediated vascular leakage was increased following loss of EC NRP1, implicating NRP1 in negative regulation of VEGFR2 signalling. In contrast, in the back skin and trachea, loss of EC NRP1 decreased vascular leakage. In accordance, phosphorylation of vascular endothelial (VE)-cadherin was increased in the ear skin but suppressed in the back skin of Nrp1 iECKO mice. NRP1 expressed on perivascular cells has been shown to impact VEGF-mediated VEGFR2 signalling. Importantly, expression of NRP1 on perivascular cells was more abundant in the ear skin than in the back skin. Global loss of NRP1 resulted in suppressed VEGFA-induced vascular leakage in the ear skin, implicating perivascular NRP1 as a juxtacrine co-receptor of VEGFA in this compartment. Altogether, we demonstrate that perivascular NRP1 is an active participant in EC VEGFA/VEGFR2 signalling and acts as an organotypic modifier of EC biology.
Collapse
Affiliation(s)
- Sagnik Pal
- Department of Immunology, Genetics and Pathology, Beijer and Science for Life Laboratories, Uppsala University, Uppsala, Sweden
| | - Yangyang Su
- Department of Immunology, Genetics and Pathology, Beijer and Science for Life Laboratories, Uppsala University, Uppsala, Sweden
| | - Emmanuel Nwadozi
- Department of Immunology, Genetics and Pathology, Beijer and Science for Life Laboratories, Uppsala University, Uppsala, Sweden
| | - Lena Claesson-Welsh
- Department of Immunology, Genetics and Pathology, Beijer and Science for Life Laboratories, Uppsala University, Uppsala, Sweden
| | - Mark Richards
- Department of Immunology, Genetics and Pathology, Beijer and Science for Life Laboratories, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
12
|
Romani P, Benedetti G, Cusan M, Arboit M, Cirillo C, Wu X, Rouni G, Kostourou V, Aragona M, Giampietro C, Grumati P, Martello G, Dupont S. Mitochondrial mechanotransduction through MIEF1 coordinates the nuclear response to forces. Nat Cell Biol 2024; 26:2046-2060. [PMID: 39433949 PMCID: PMC11628398 DOI: 10.1038/s41556-024-01527-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 09/12/2024] [Indexed: 10/23/2024]
Abstract
Tissue-scale architecture and mechanical properties instruct cell behaviour under physiological and diseased conditions, but our understanding of the underlying mechanisms remains fragmentary. Here we show that extracellular matrix stiffness, spatial confinements and applied forces, including stretching of mouse skin, regulate mitochondrial dynamics. Actomyosin tension promotes the phosphorylation of mitochondrial elongation factor 1 (MIEF1), limiting the recruitment of dynamin-related protein 1 (DRP1) at mitochondria, as well as peri-mitochondrial F-actin formation and mitochondrial fission. Strikingly, mitochondrial fission is also a general mechanotransduction mechanism. Indeed, we found that DRP1- and MIEF1/2-dependent fission is required and sufficient to regulate three transcription factors of broad relevance-YAP/TAZ, SREBP1/2 and NRF2-to control cell proliferation, lipogenesis, antioxidant metabolism, chemotherapy resistance and adipocyte differentiation in response to mechanical cues. This extends to the mouse liver, where DRP1 regulates hepatocyte proliferation and identity-hallmark YAP-dependent phenotypes. We propose that mitochondria fulfil a unifying signalling function by which the mechanical tissue microenvironment coordinates complementary cell functions.
Collapse
Affiliation(s)
- Patrizia Romani
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Giada Benedetti
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Martina Cusan
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Mattia Arboit
- Department of Biology, University of Padova, Padova, Italy
| | - Carmine Cirillo
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | - Xi Wu
- Department of Mechanical and Process Engineering, ETH Zurich, Zurich, Switzerland
| | - Georgia Rouni
- Institute for Bioinnovation, Biomedical Sciences Research Centre "Alexander Fleming", Athens, Greece
| | - Vassiliki Kostourou
- Institute for Bioinnovation, Biomedical Sciences Research Centre "Alexander Fleming", Athens, Greece
| | - Mariaceleste Aragona
- Novo Nordisk Foundation Center for Stem Cell Medicine (ReNEW), University of Copenhagen, Copenhagen, Denmark
| | - Costanza Giampietro
- Department of Mechanical and Process Engineering, ETH Zurich, Zurich, Switzerland
- Swiss Federal Laboratories for Materials Science and Technology, Dübendorf, Switzerland
| | - Paolo Grumati
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | | | - Sirio Dupont
- Department of Molecular Medicine, University of Padova, Padova, Italy.
| |
Collapse
|
13
|
Lin YC, Chang YJ, Gau SS, Lo CM, Yang RB. SCUBE2 regulates adherens junction dynamics and vascular barrier function during inflammation. Cardiovasc Res 2024; 120:1636-1649. [PMID: 38870316 DOI: 10.1093/cvr/cvae132] [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: 09/20/2023] [Revised: 01/22/2024] [Accepted: 05/03/2024] [Indexed: 06/15/2024] Open
Abstract
AIMS SCUBE2 (signal peptide-CUB-epidermal growth factor-like domain-containing protein 2) is a secreted or membrane-bound protein originally identified from endothelial cells (ECs). Our previous work showed that SCUBE2 forms a complex with E-cadherin and stabilizes epithelial adherens junctions (AJs) to promote epithelial phenotypes. However, it remains unclear whether SCUBE2 also interacts with vascular endothelial (VE)-cadherin and modulates EC barrier function. In this study, we investigated whether and how SCUBE2 in ECs regulates vascular barrier maintenance. METHODS AND RESULTS We showed that SCUBE2 colocalized and interacted with VE-cadherin and VE-protein tyrosine phosphatase (VE-PTP) within EC AJs. Furthermore, SCUBE2 knockdown disrupted EC AJs and increased EC permeability. Expression of EC SCUBE2 was suppressed at both mRNA and protein levels via the nuclear factor-κB signalling pathway in response to pro-inflammatory cytokines or permeability-inducing agents. In line with these findings, EC-specific deletion of Scube2 (EC-KO) in mice impaired baseline barrier function and worsened vascular leakiness of peripheral capillaries after local injection of histamine or vascular endothelial growth factor. EC-KO mice were also sensitive to pulmonary vascular hyperpermeability and leucocyte infiltration in response to acute endotoxin- or influenza virus-induced systemic inflammation. Meanwhile, EC-specific SCUBE2-overexpressing mice were protected from these effects. Molecular studies suggested that SCUBE2 acts as a scaffold molecule enabling VE-PTP to dephosphorylate VE-cadherin, which prevents VE-cadherin internalization and stabilizes EC AJs. As such, loss of SCUBE2 resulted in hyperphosphorylation of VE-cadherin at tyrosine 685, which led to its endocytosis, thus destabilizing EC AJs and reducing barrier function. All of these effects were exacerbated by inflammatory insults. CONCLUSION We found that SCUBE2 contributes to vascular integrity by recruiting VE-PTP to dephosphorylate VE-cadherin and stabilize AJs, thereby promoting EC barrier function. Moreover, our data suggest that genetic overexpression or pharmacological up-regulation of SCUBE2 may help to prevent vascular leakage and oedema in inflammatory diseases.
Collapse
Affiliation(s)
- Yuh-Charn Lin
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
| | - Ya-Jen Chang
- Institute of Biomedical Sciences, Academia Sinica, 128 Academia Rd., Sec. 2, Taipei 115201, Taiwan
| | - Shiang-Shin Gau
- Institute of Biomedical Sciences, Academia Sinica, 128 Academia Rd., Sec. 2, Taipei 115201, Taiwan
- Graduate School of Biostudies, Kyoto University, Kyoto 6068501, Japan
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 6068501, Japan
| | - Chun-Min Lo
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Ruey-Bing Yang
- Institute of Biomedical Sciences, Academia Sinica, 128 Academia Rd., Sec. 2, Taipei 115201, Taiwan
- Biomedical Translation Research Center, Academia Sinica, 99, Ln. 130, Academia Rd., Sec. 1, Taipei 115201, Taiwan
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, 250 Wuxing St., Taipei 110301, Taiwan
| |
Collapse
|
14
|
Johnson D, Colijn S, Richee J, Yano J, Burns M, Davis AE, Pham VN, Saric A, Jain A, Yin Y, Castranova D, Melani M, Fujita M, Grainger S, Bonifacino JS, Weinstein BM, Stratman AN. Angiogenesis is limited by LIC1-mediated lysosomal trafficking. Angiogenesis 2024; 27:943-962. [PMID: 39356418 PMCID: PMC11653708 DOI: 10.1007/s10456-024-09951-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 09/25/2024] [Indexed: 10/03/2024]
Abstract
Dynein cytoplasmic 1 light intermediate chain 1 (LIC1, DYNC1LI1) is a core subunit of the dynein motor complex. The LIC1 subunit also interacts with various cargo adaptors to regulate Rab-mediated endosomal recycling and lysosomal degradation. Defects in this gene are predicted to alter dynein motor function, Rab binding capabilities, and cytoplasmic cargo trafficking. Here, we have identified a dync1li1 zebrafish mutant, harboring a premature stop codon at the exon 12/13 splice acceptor site, that displays increased angiogenesis. In vitro, LIC1-deficient human endothelial cells display increases in cell surface levels of the pro-angiogenic receptor VEGFR2, SRC phosphorylation, and Rab11-mediated endosomal recycling. In vivo, endothelial-specific expression of constitutively active Rab11a leads to excessive angiogenesis, similar to the dync1li1 mutants. Increased angiogenesis is also evident in zebrafish harboring mutations in rilpl1/2, the adaptor proteins that promote Rab docking to Lic1 to mediate lysosomal targeting. These findings suggest that LIC1 and the Rab-adaptor proteins RILPL1 and 2 restrict angiogenesis by promoting degradation of VEGFR2-containing recycling endosomes. Disruption of LIC1- and RILPL1/2-mediated lysosomal targeting increases Rab11-mediated recycling endosome activity, promoting excessive SRC signaling and angiogenesis.
Collapse
Affiliation(s)
- Dymonn Johnson
- Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO, 63110, USA
| | - Sarah Colijn
- Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO, 63110, USA
| | - Jahmiera Richee
- Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO, 63110, USA
| | - Joseph Yano
- Division of Developmental Biology, Section on Vertebrate Organogenesis, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
- Cell and Molecular Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Margaret Burns
- Division of Developmental Biology, Section on Vertebrate Organogenesis, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Andrew E Davis
- Division of Developmental Biology, Section on Vertebrate Organogenesis, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Van N Pham
- Division of Developmental Biology, Section on Vertebrate Organogenesis, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Amra Saric
- Section On Intracellular Protein Trafficking, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
- Neurosciences and Mental Health Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
| | - Akansha Jain
- Section On Intracellular Protein Trafficking, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ying Yin
- Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO, 63110, USA
| | - Daniel Castranova
- Division of Developmental Biology, Section on Vertebrate Organogenesis, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Mariana Melani
- Division of Developmental Biology, Section on Vertebrate Organogenesis, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
- Fundación Instituto Leloir, Buenos Aires, Argentina
- Consejo Nacional De Investigaciones Científicas Y Técnicas (CONICET), Buenos Aires, Argentina
- Departamento De Fisiología, Biología Molecular Y Celular, Facultad De Ciencias Exactas Y Naturales, Universidad De Buenos Aires, Buenos Aires, Argentina
| | - Misato Fujita
- Division of Developmental Biology, Section on Vertebrate Organogenesis, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
- Kanagawa University, Kanagawa, 221-8686, Japan
| | - Stephanie Grainger
- Department of Cell Biology, Van Andel Institute, Grand Rapids, MI, 49503, USA
| | - Juan S Bonifacino
- Section On Intracellular Protein Trafficking, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Brant M Weinstein
- Division of Developmental Biology, Section on Vertebrate Organogenesis, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Amber N Stratman
- Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
15
|
Liao YL, Fang YF, Sun JX, Dou GR. Senescent endothelial cells: a potential target for diabetic retinopathy. Angiogenesis 2024; 27:663-679. [PMID: 39215875 PMCID: PMC11564237 DOI: 10.1007/s10456-024-09943-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
Diabetic retinopathy (DR) is a diabetic complication that results in visual impairment and relevant retinal diseases. Current therapeutic strategies on DR primarily focus on antiangiogenic therapies, which particularly target vascular endothelial growth factor and its related signaling transduction. However, these therapies still have limitations due to the intricate pathogenesis of DR. Emerging studies have shown that premature senescence of endothelial cells (ECs) in a hyperglycemic environment is involved in the disease process of DR and plays multiple roles at different stages. Moreover, these surprising discoveries have driven the development of senotherapeutics and strategies targeting senescent endothelial cells (SECs), which present challenging but promising prospects in DR treatment. In this review, we focus on the inducers and mechanisms of EC senescence in the pathogenesis of DR and summarize the current research advances in the development of senotherapeutics and strategies that target SECs for DR treatment. Herein, we highlight the role played by key factors at different stages of EC senescence, which will be critical for facilitating the development of future innovative treatment strategies that target the different stages of senescence in DR.
Collapse
Affiliation(s)
- Ying-Lu Liao
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
- Department of the Cadet Team 6 of the School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Yi-Fan Fang
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Jia-Xing Sun
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Guo-Rui Dou
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
16
|
Stegmeyer RI, Holstein K, Spring K, Timmerman I, Xia M, Stasch M, Möller T, Nottebaum AF, Vestweber D. Csk controls leukocyte extravasation via local regulation of Src family kinases and cortactin signaling. Front Immunol 2024; 15:1480152. [PMID: 39530094 PMCID: PMC11550946 DOI: 10.3389/fimmu.2024.1480152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024] Open
Abstract
C-terminal Src kinase (Csk) targets Src family kinases (SFKs) and thereby inactivates them. We have previously shown that Csk binds to phosphorylated tyrosine 685 of VE-cadherin, an adhesion molecule of major importance for the regulation of endothelial junctions. This tyrosine residue is an SFK target, and its mutation (VE-cadherin-Y685F) inhibits the induction of vascular permeability in various inflammation models. Nevertheless, surprisingly, it increases leukocyte extravasation. Here, we investigated whether endothelial Csk is involved in these effects. We found that the deficiency of Csk in endothelial cells augments SFK activation and the phosphorylation of VE-cadherin-Y685 but had no net effect on vascular leak formation. In contrast, the lack of endothelial Csk enhanced leukocyte adhesion and transmigration in vitro and in vivo. Furthermore, the silencing of Csk increased tyrosine phosphorylation of the SFK substrate cortactin. Importantly, the effects of Csk silencing on the increase in SFK activation, cortactin phosphorylation, and neutrophil diapedesis were all dependent on Y685 of VE-cadherin. Deletion of cortactin, in turn, erased the supporting effect of Csk silencing on leukocyte transmigration. We have previously shown that leukocyte transmigration is regulated by endothelial cortactin in an ICAM-1-dependent manner. In line with this, blocking of ICAM-1 erased the supporting effect of Csk silencing on leukocyte transmigration. Collectively, our results establish a negative feedback loop that depends on the phosphorylation of VE-cadherin-Y685, which recruits Csk, which in turn dampens the activation of SFK and cortactin and thereby the clustering of ICAM-1 and the extravasation of neutrophils.
Collapse
Affiliation(s)
- Rebekka I. Stegmeyer
- Department of Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Katrin Holstein
- Department of Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Kathleen Spring
- Department of Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Ilse Timmerman
- Department of Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Min Xia
- Department of Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Malte Stasch
- BioOptic Service, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Tanja Möller
- Department of Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Astrid F. Nottebaum
- Department of Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Dietmar Vestweber
- Department of Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| |
Collapse
|
17
|
Ouyang P, Cheng B, He X, Lou J, Li X, Guo H, Xu F. Navigating the biophysical landscape: how physical cues steer the journey of bone metastatic tumor cells. Trends Cancer 2024; 10:792-808. [PMID: 39127608 DOI: 10.1016/j.trecan.2024.07.003] [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: 03/29/2024] [Revised: 07/08/2024] [Accepted: 07/12/2024] [Indexed: 08/12/2024]
Abstract
Many tumors prefer to metastasize to bone, but the underlying mechanisms remain elusive. The human skeletal system has unique physical properties, that are distinct from other organs, which play a key role in directing the behavior of tumor cells within bone. Understanding the physical journey of tumor cells within bone is crucial. In this review we discuss bone metastasis in the context of how physical cues in the bone vasculature and bone marrow niche regulate the fate of tumor cells. Our objective is to inspire innovative diagnostic and therapeutic approaches for bone metastasis from a mechanobiological perspective.
Collapse
Affiliation(s)
- Pengrong Ouyang
- Department of Orthopedic Surgery, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, P.R. China; Bioinspired Engineering and Biomechanics Center (BEBC), Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P.R. China
| | - Bo Cheng
- Bioinspired Engineering and Biomechanics Center (BEBC), Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P.R. China; TFX Group-Xi'an Jiaotong University Institute of Life Health, Xi'an 710049, P.R. China
| | - Xijing He
- Department of Orthopedic Surgery, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, P.R. China; Xi'an International Medical Center Hospital, Xi'an 710061, P.R. China.
| | - Jiatao Lou
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China.
| | - Xiaokang Li
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, P.R. China.
| | - Hui Guo
- Department of Medical Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, P.R. China.
| | - Feng Xu
- Bioinspired Engineering and Biomechanics Center (BEBC), Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P.R. China.
| |
Collapse
|
18
|
Wilkens M, Holtermann L, Stahl AK, Stegmeyer RI, Nottebaum AF, Vestweber D. Ubiquitination of VE-cadherin regulates inflammation-induced vascular permeability in vivo. EMBO Rep 2024; 25:4013-4032. [PMID: 39112792 PMCID: PMC11387630 DOI: 10.1038/s44319-024-00221-7] [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: 11/22/2023] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 09/12/2024] Open
Abstract
VE-cadherin is a major component of the cell adhesion machinery which provides integrity and plasticity of the barrier function of endothelial junctions. Here, we analyze whether ubiquitination of VE-cadherin is involved in the regulation of the endothelial barrier in inflammation in vivo. We show that histamine and thrombin stimulate ubiquitination of VE-cadherin in HUVEC, which is completely blocked if the two lysine residues K626 and K633 are replaced by arginine. Similarly, these mutations block histamine-induced endocytosis of VE-cadherin. We describe two knock-in mouse lines with endogenous VE-cadherin being replaced by either a VE-cadherin K626/633R or a VE-cadherin KallR mutant, where all seven lysine residues are mutated. Mutant mice are viable, healthy and fertile with normal expression levels of junctional VE-cadherin. Histamine- or LPS-induced vascular permeability in the skin or lung of both of these mutant mice are clearly and similarly reduced in comparison to WT mice. Additionally, we detect a role of K626/633 for lysosomal targeting. Collectively, our findings identify ubiquitination of VE-cadherin as important for the induction of vascular permeability in the inflamed skin and lung.
Collapse
Affiliation(s)
- Markus Wilkens
- Max Planck Institute for Molecular Biomedicine, D-48149, Muenster, Germany
| | - Leonie Holtermann
- Max Planck Institute for Molecular Biomedicine, D-48149, Muenster, Germany
| | - Ann-Kathrin Stahl
- Max Planck Institute for Molecular Biomedicine, D-48149, Muenster, Germany
| | | | - Astrid F Nottebaum
- Max Planck Institute for Molecular Biomedicine, D-48149, Muenster, Germany
| | - Dietmar Vestweber
- Max Planck Institute for Molecular Biomedicine, D-48149, Muenster, Germany.
| |
Collapse
|
19
|
Jeong JY, Bafor AE, Freeman BH, Chen PR, Park ES, Kim E. Pathophysiology in Brain Arteriovenous Malformations: Focus on Endothelial Dysfunctions and Endothelial-to-Mesenchymal Transition. Biomedicines 2024; 12:1795. [PMID: 39200259 PMCID: PMC11351371 DOI: 10.3390/biomedicines12081795] [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: 06/26/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 09/02/2024] Open
Abstract
Brain arteriovenous malformations (bAVMs) substantially increase the risk for intracerebral hemorrhage (ICH), which is associated with significant morbidity and mortality. However, the treatment options for bAVMs are severely limited, primarily relying on invasive methods that carry their own risks for intraoperative hemorrhage or even death. Currently, there are no pharmaceutical agents shown to treat this condition, primarily due to a poor understanding of bAVM pathophysiology. For the last decade, bAVM research has made significant advances, including the identification of novel genetic mutations and relevant signaling in bAVM development. However, bAVM pathophysiology is still largely unclear. Further investigation is required to understand the detailed cellular and molecular mechanisms involved, which will enable the development of safer and more effective treatment options. Endothelial cells (ECs), the cells that line the vascular lumen, are integral to the pathogenesis of bAVMs. Understanding the fundamental role of ECs in pathological conditions is crucial to unraveling bAVM pathophysiology. This review focuses on the current knowledge of bAVM-relevant signaling pathways and dysfunctions in ECs, particularly the endothelial-to-mesenchymal transition (EndMT).
Collapse
Affiliation(s)
| | | | | | | | | | - Eunhee Kim
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (J.Y.J.); (A.E.B.); (B.H.F.); (P.R.C.); (E.S.P.)
| |
Collapse
|
20
|
Shan KZ, Le T, Liang P, Dong P, Lowry AJ, Kremmyda P, Claesson-Welsh L, Yang H. TMEM16F scramblase regulates angiogenesis via endothelial intracellular signaling. J Cell Sci 2024; 137:jcs261566. [PMID: 38940198 PMCID: PMC11273297 DOI: 10.1242/jcs.261566] [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: 08/17/2023] [Accepted: 06/19/2024] [Indexed: 06/29/2024] Open
Abstract
TMEM16F (also known as ANO6), a Ca2+-activated lipid scramblase (CaPLSase) that dynamically disrupts lipid asymmetry, plays a crucial role in various physiological and pathological processes, such as blood coagulation, neurodegeneration, cell-cell fusion and viral infection. However, the mechanisms through which it regulates these processes remain largely elusive. Using endothelial cell-mediated angiogenesis as a model, here we report a previously unknown intracellular signaling function of TMEM16F. We demonstrate that TMEM16F deficiency impairs developmental retinal angiogenesis in mice and disrupts angiogenic processes in vitro. Biochemical analyses indicate that the absence of TMEM16F enhances the plasma membrane association of activated Src kinase. This in turn increases VE-cadherin phosphorylation and downregulation, accompanied by suppressed angiogenesis. Our findings not only highlight the role of intracellular signaling by TMEM16F in endothelial cells but also open new avenues for exploring the regulatory mechanisms for membrane lipid asymmetry and their implications in disease pathogenesis.
Collapse
Affiliation(s)
- Ke Zoe Shan
- Department of Biochemistry, Duke University, School of Medicine, Durham, NC 27710, USA
| | - Trieu Le
- Department of Biochemistry, Duke University, School of Medicine, Durham, NC 27710, USA
| | - Pengfei Liang
- Department of Biochemistry, Duke University, School of Medicine, Durham, NC 27710, USA
| | - Ping Dong
- Department of Biochemistry, Duke University, School of Medicine, Durham, NC 27710, USA
| | - Augustus J. Lowry
- Department of Biochemistry, Duke University, School of Medicine, Durham, NC 27710, USA
| | - Polina Kremmyda
- Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck, Beijer and SciLifeLab Laboratory, Uppsala 751 85, Sweden
| | - Lena Claesson-Welsh
- Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck, Beijer and SciLifeLab Laboratory, Uppsala 751 85, Sweden
| | - Huanghe Yang
- Department of Biochemistry, Duke University, School of Medicine, Durham, NC 27710, USA
- Department of Neurobiology, Duke University, School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
21
|
Essebier P, Keyser M, Yordanov T, Hill B, Yu A, Noordstra I, Yap AS, Stehbens SJ, Lagendijk AK, Schimmel L, Gordon EJ. c-Src-induced vascular malformations require localised matrix degradation at focal adhesions. J Cell Sci 2024; 137:jcs262101. [PMID: 38881365 PMCID: PMC11267457 DOI: 10.1242/jcs.262101] [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: 03/07/2024] [Accepted: 06/06/2024] [Indexed: 06/18/2024] Open
Abstract
Endothelial cells lining the blood vessel wall communicate intricately with the surrounding extracellular matrix, translating mechanical cues into biochemical signals. Moreover, vessels require the capability to enzymatically degrade the matrix surrounding them, to facilitate vascular expansion. c-Src plays a key role in blood vessel growth, with its loss in the endothelium reducing vessel sprouting and focal adhesion signalling. Here, we show that constitutive activation of c-Src in endothelial cells results in rapid vascular expansion, operating independently of growth factor stimulation or fluid shear stress forces. This is driven by an increase in focal adhesion signalling and size, with enhancement of localised secretion of matrix metalloproteinases responsible for extracellular matrix remodelling. Inhibition of matrix metalloproteinase activity results in a robust rescue of the vascular expansion elicited by heightened c-Src activity. This supports the premise that moderating focal adhesion-related events and matrix degradation can counteract abnormal vascular expansion, with implications for pathologies driven by unusual vascular morphologies.
Collapse
Affiliation(s)
- Patricia Essebier
- Centre for Cell Biology of Chronic Disease, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia4072
| | - Mikaela Keyser
- Centre for Cell Biology of Chronic Disease, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia4072
| | - Teodor Yordanov
- Centre for Cell Biology of Chronic Disease, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia4072
| | - Brittany Hill
- Centre for Cell Biology of Chronic Disease, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia4072
| | - Alexander Yu
- Centre for Cell Biology of Chronic Disease, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia4072
| | - Ivar Noordstra
- Centre for Cell Biology of Chronic Disease, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia4072
| | - Alpha S. Yap
- Centre for Cell Biology of Chronic Disease, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia4072
| | - Samantha J. Stehbens
- Centre for Cell Biology of Chronic Disease, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia4072
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia4072
| | - Anne K. Lagendijk
- Centre for Cell Biology of Chronic Disease, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia4072
| | - Lilian Schimmel
- Centre for Cell Biology of Chronic Disease, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia4072
| | - Emma J. Gordon
- Centre for Cell Biology of Chronic Disease, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia4072
| |
Collapse
|
22
|
Wakasugi R, Suzuki K, Kaneko-Kawano T. Molecular Mechanisms Regulating Vascular Endothelial Permeability. Int J Mol Sci 2024; 25:6415. [PMID: 38928121 PMCID: PMC11203514 DOI: 10.3390/ijms25126415] [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: 04/30/2024] [Revised: 05/30/2024] [Accepted: 06/02/2024] [Indexed: 06/28/2024] Open
Abstract
Vascular endothelial cells form a monolayer in the vascular lumen and act as a selective barrier to control the permeability between blood and tissues. To maintain homeostasis, the endothelial barrier function must be strictly integrated. During acute inflammation, vascular permeability temporarily increases, allowing intravascular fluid, cells, and other components to permeate tissues. Moreover, it has been suggested that the dysregulation of endothelial cell permeability may cause several diseases, including edema, cancer, and atherosclerosis. Here, we reviewed the molecular mechanisms by which endothelial cells regulate the barrier function and physiological permeability.
Collapse
Affiliation(s)
| | | | - Takako Kaneko-Kawano
- Graduate School of Pharmacy, Ritsumeikan University, 1-1-1 Noji-higashi, Kusatsu 525-8577, Shiga, Japan; (R.W.); (K.S.)
| |
Collapse
|
23
|
Burg N, Malpass R, Alex L, Tran M, Englebrecht E, Kuo A, Pannelini T, Minett M, Athukorala K, Worgall T, Faust HJ, Goodman S, Mehta B, Brenner M, Vestweber D, Wei K, Blobel C, Hla T, Salmon JE. Endothelial cell sphingosine 1-phosphate receptor 1 restrains VE-cadherin cleavage and attenuates experimental inflammatory arthritis. JCI Insight 2024; 9:e171467. [PMID: 38855867 PMCID: PMC11382883 DOI: 10.1172/jci.insight.171467] [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: 06/06/2023] [Accepted: 05/01/2024] [Indexed: 06/11/2024] Open
Abstract
In rheumatoid arthritis, inflammatory mediators extravasate from blood into joints via gaps between endothelial cells (ECs), but the contribution of ECs is not known. Sphingosine 1-phosphate receptor 1 (S1PR1), widely expressed on ECs, maintains the vascular barrier. Here, we assessed the contribution of vascular integrity and EC S1PR1 signaling to joint damage in mice exposed to serum-induced arthritis (SIA). EC-specific deletion of S1PR1 or pharmacological blockade of S1PR1 promoted vascular leak and amplified SIA, whereas overexpression of EC S1PR1 or treatment with an S1PR1 agonist delayed SIA. Blockade of EC S1PR1 induced membrane metalloproteinase-dependent cleavage of vascular endothelial cadherin (VE-cadherin), a principal adhesion molecule that maintains EC junctional integrity. We identified a disintegrin and a metalloproteinase domain 10 (ADAM10) as the principal VE-cadherin "sheddase." Mice expressing a stabilized VE-cadherin construct had decreased extravascular VE-cadherin and vascular leakage in response to S1PR1 blockade, and they were protected from SIA. Importantly, patients with active rheumatoid arthritis had decreased circulating S1P and microvascular expression of S1PR1, suggesting a dysregulated S1P/S1PR1 axis favoring vascular permeability and vulnerability. We present a model in which EC S1PR1 signaling maintains homeostatic vascular barrier function by limiting VE-cadherin shedding mediated by ADAM10 and suggest this signaling axis as a therapeutic target in inflammatory arthritis.
Collapse
Affiliation(s)
- Nathalie Burg
- Hospital for Special Surgery, New York, New York, USA
| | - Ryan Malpass
- Hospital for Special Surgery, New York, New York, USA
| | - Linda Alex
- Hospital for Special Surgery, New York, New York, USA
| | - Miles Tran
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Eric Englebrecht
- School of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Andrew Kuo
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | - Tilla Worgall
- Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Heather J. Faust
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Susan Goodman
- Hospital for Special Surgery, New York, New York, USA
| | - Bella Mehta
- Hospital for Special Surgery, New York, New York, USA
| | - Michael Brenner
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | - Kevin Wei
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Carl Blobel
- Hospital for Special Surgery, New York, New York, USA
| | - Timothy Hla
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
24
|
Teuwen JTJ, van der Vorst EPC, Maas SL. Navigating the Maze of Kinases: CaMK-like Family Protein Kinases and Their Role in Atherosclerosis. Int J Mol Sci 2024; 25:6213. [PMID: 38892400 PMCID: PMC11172518 DOI: 10.3390/ijms25116213] [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: 04/13/2024] [Revised: 05/30/2024] [Accepted: 06/02/2024] [Indexed: 06/21/2024] Open
Abstract
Circulating low-density lipoprotein (LDL) levels are a major risk factor for cardiovascular diseases (CVD), and even though current treatment strategies focusing on lowering lipid levels are effective, CVD remains the primary cause of death worldwide. Atherosclerosis is the major cause of CVD and is a chronic inflammatory condition in which various cell types and protein kinases play a crucial role. However, the underlying mechanisms of atherosclerosis are not entirely understood yet. Notably, protein kinases are highly druggable targets and represent, therefore, a novel way to target atherosclerosis. In this review, the potential role of the calcium/calmodulin-dependent protein kinase-like (CaMKL) family and its role in atherosclerosis will be discussed. This family consists of 12 subfamilies, among which are the well-described and conserved liver kinase B1 (LKB1) and 5' adenosine monophosphate-activated protein kinase (AMPK) subfamilies. Interestingly, LKB1 plays a key role and is considered a master kinase within the CaMKL family. It has been shown that LKB1 signaling leads to atheroprotective effects, while, for example, members of the microtubule affinity-regulating kinase (MARK) subfamily have been described to aggravate atherosclerosis development. These observations highlight the importance of studying kinases and their signaling pathways in atherosclerosis, bringing us a step closer to unraveling the underlying mechanisms of atherosclerosis.
Collapse
Affiliation(s)
- Jules T. J. Teuwen
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany;
- Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074 Aachen, Germany
| | - Emiel P. C. van der Vorst
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany;
- Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074 Aachen, Germany
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52074 Aachen, Germany
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität München, 80336 München, Germany
| | - Sanne L. Maas
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany;
- Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074 Aachen, Germany
| |
Collapse
|
25
|
Weinstein N, Carlsen J, Schulz S, Stapleton T, Henriksen HH, Travnik E, Johansson PI. A Lifelike guided journey through the pathophysiology of pulmonary hypertension-from measured metabolites to the mechanism of action of drugs. Front Cardiovasc Med 2024; 11:1341145. [PMID: 38845688 PMCID: PMC11153715 DOI: 10.3389/fcvm.2024.1341145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 04/12/2024] [Indexed: 06/09/2024] Open
Abstract
Introduction Pulmonary hypertension (PH) is a pathological condition that affects approximately 1% of the population. The prognosis for many patients is poor, even after treatment. Our knowledge about the pathophysiological mechanisms that cause or are involved in the progression of PH is incomplete. Additionally, the mechanism of action of many drugs used to treat pulmonary hypertension, including sotatercept, requires elucidation. Methods Using our graph-powered knowledge mining software Lifelike in combination with a very small patient metabolite data set, we demonstrate how we derive detailed mechanistic hypotheses on the mechanisms of PH pathophysiology and clinical drugs. Results In PH patients, the concentration of hypoxanthine, 12(S)-HETE, glutamic acid, and sphingosine 1 phosphate is significantly higher, while the concentration of L-arginine and L-histidine is lower than in healthy controls. Using the graph-based data analysis, gene ontology, and semantic association capabilities of Lifelike, led us to connect the differentially expressed metabolites with G-protein signaling and SRC. Then, we associated SRC with IL6 signaling. Subsequently, we found associations that connect SRC, and IL6 to activin and BMP signaling. Lastly, we analyzed the mechanisms of action of several existing and novel pharmacological treatments for PH. Lifelike elucidated the interplay between G-protein, IL6, activin, and BMP signaling. Those pathways regulate hallmark pathophysiological processes of PH, including vasoconstriction, endothelial barrier function, cell proliferation, and apoptosis. Discussion The results highlight the importance of SRC, ERK1, AKT, and MLC activity in PH. The molecular pathways affected by existing and novel treatments for PH also converge on these molecules. Importantly, sotatercept affects SRC, ERK1, AKT, and MLC simultaneously. The present study shows the power of mining knowledge graphs using Lifelike's diverse set of data analytics functionalities for developing knowledge-driven hypotheses on PH pathophysiological and drug mechanisms and their interactions. We believe that Lifelike and our presented approach will be valuable for future mechanistic studies of PH, other diseases, and drugs.
Collapse
Affiliation(s)
- Nathan Weinstein
- CAG Center for Endotheliomics, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Jørn Carlsen
- CAG Center for Endotheliomics, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Sebastian Schulz
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Timothy Stapleton
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Hanne H. Henriksen
- CAG Center for Endotheliomics, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Evelyn Travnik
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Pär Ingemar Johansson
- CAG Center for Endotheliomics, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
26
|
Chen-Li G, Martinez-Archer R, Coghi A, Roca JA, Rodriguez FJ, Acaba-Berrocal L, Berrocal MH, Wu L. Beyond VEGF: Angiopoietin-Tie Signaling Pathway in Diabetic Retinopathy. J Clin Med 2024; 13:2778. [PMID: 38792322 PMCID: PMC11122151 DOI: 10.3390/jcm13102778] [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: 02/26/2024] [Revised: 04/12/2024] [Accepted: 04/30/2024] [Indexed: 05/26/2024] Open
Abstract
Complications from diabetic retinopathy such as diabetic macular edema (DME) and proliferative diabetic retinopathy (PDR) constitute leading causes of preventable vision loss in working-age patients. Since vascular endothelial growth factor (VEGF) plays a major role in the pathogenesis of these complications, VEGF inhibitors have been the cornerstone of their treatment. Anti-VEGF monotherapy is an effective but burdensome treatment for DME. However, due to the intensive and burdensome treatment, most patients in routine clinical practice are undertreated, and therefore, their outcomes are compromised. Even in adequately treated patients, persistent DME is reported anywhere from 30% to 60% depending on the drug used. PDR is currently treated by anti-VEGF, panretinal photocoagulation (PRP) or a combination of both. Similarly, a number of eyes, despite these treatments, continue to progress to tractional retinal detachment and vitreous hemorrhage. Clearly there are other molecular pathways other than VEGF involved in the pathogenesis of DME and PDR. One of these pathways is the angiopoietin-Tie signaling pathway. Angiopoietin 1 (Ang1) plays a major role in maintaining vascular quiescence and stability. It acts as a molecular brake against vascular destabilization and inflammation that is usually promoted by angiopoietin 2 (Ang2). Several pathological conditions including chronic hyperglycemia lead to Ang2 upregulation. Recent regulatory approval of the bi-specific antibody, faricimab, may improve long term outcomes in DME. It targets both the Ang/Tie and VEGF pathways. The YOSEMITE and RHINE were multicenter, double-masked, randomized non-inferiority phase 3 clinical trials that compared faricimab to aflibercept in eyes with center-involved DME. At 12 months of follow-up, faricimab demonstrated non-inferior vision gains, improved anatomic outcomes and a potential for extended dosing when compared to aflibercept. The 2-year results of the YOSEMITE and RHINE trials demonstrated that the anatomic and functional results obtained at the 1 year follow-up were maintained. Short term outcomes of previously treated and treatment-naive eyes with DME that were treated with faricimab during routine clinical practice suggest a beneficial effect of faricimab over other agents. Targeting of Ang2 has been reported by several other means including VE-PTP inhibitors, integrin binding peptide and surrobodies.
Collapse
Affiliation(s)
- Genesis Chen-Li
- Asociados de Mácula Vitreo y Retina de Costa Rica, San José 60612, Costa Rica (R.M.-A.); (A.C.)
| | - Rebeca Martinez-Archer
- Asociados de Mácula Vitreo y Retina de Costa Rica, San José 60612, Costa Rica (R.M.-A.); (A.C.)
| | - Andres Coghi
- Asociados de Mácula Vitreo y Retina de Costa Rica, San José 60612, Costa Rica (R.M.-A.); (A.C.)
| | | | | | - Luis Acaba-Berrocal
- Department of Ophthalmology, Illinois Eye and Ear Infirmary, School of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | | | - Lihteh Wu
- Asociados de Mácula Vitreo y Retina de Costa Rica, San José 60612, Costa Rica (R.M.-A.); (A.C.)
- Department of Ophthalmology, Illinois Eye and Ear Infirmary, School of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| |
Collapse
|
27
|
Lai Y, Yang N, Chen X, Ma X, Chen Z, Dong C, Yu G, Huang Y, Shi D, Fang P, Fu K, Jiang R, Mao C, Ding J, Gao W. Dihydrocapsaicin suppresses the STING-mediated accumulation of ROS and NLRP3 inflammasome and alleviates apoptosis after ischemia-reperfusion injury of perforator skin flap. Phytother Res 2024; 38:2539-2559. [PMID: 38459660 DOI: 10.1002/ptr.8167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 01/16/2024] [Accepted: 02/08/2024] [Indexed: 03/10/2024]
Abstract
Avascular necrosis frequently occurs as a complication following surgery involving the distal perforator flap. Dihydrocapsaicin (DHC) can protect tissue from ischemia-reperfusion (I/R) injury, but its specific role in multizone perforator flaps remains unclear. In this study, the prospective target of DHC in the context of I/R injury was predicted using network pharmacology analysis. Flap viability was determined through survival area analysis, laser Doppler blood flow, angiograms, and histological examination. The expressions of angiogenesis, apoptosis, NLR family pyrin domain containing 3 (NLRP3) inflammasome, oxidative stress, and molecules related to cyclic guanosine monophosphate (GMP)-adenosine monophosphate synthase (cGAS)-interferon gene stimulant (STING) pathway were assessed using western blotting, immunofluorescence, TUNEL staining, and dihydroethidium (DHE) staining. Our finding revealed that DHC promoted the perforator flap survival, which involves the cGAS-STING pathway, oxidative stress, NLRP3 inflammasome, apoptosis, and angiogenesis. DHC induced oxidative stress resistance and suppressed the NLRP3 inflammasome, preventing apoptosis in vascular endothelial cells. Through regulation of STING pathway, DHC controlled oxidative stress in endothelial cells and NLRP3 levels in ischemic flaps. However, activation of the cGAS-STING pathway led to the accumulation of reactive oxygen species (ROS) and NLRP3 inflammasome, thereby diminishing the protective role of DHC. DHC enhanced the survival of multidomain perforator flaps by suppressing the cGAS-STING pathway, oxidative stress, and the formation of NLRP3 inflammasome. These findings unveil a potentially novel mechanism with clinical significance for promoting the survival of multidomain perforator flaps.
Collapse
Affiliation(s)
- Yingying Lai
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Ningning Yang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Xuankuai Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Xianhui Ma
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Zhuliu Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Chengji Dong
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Gaoxiang Yu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Yingying Huang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Donghao Shi
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Pin Fang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Kejian Fu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Renhao Jiang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Cong Mao
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Jian Ding
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Weiyang Gao
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
28
|
Wen Z, Fan J, Zhan F, Li X, Li B, Lu P, Yao X, Shen Z, Liu Z, Wang C, Li X, Jin W, Zhang X, Qi Y, Wang X, Song M. The role of FPR2-mediated ferroptosis in formyl peptide-induced acute lung injury against endothelial barrier damage and protective effect of the mitochondria-derived peptide MOTS-c. Int Immunopharmacol 2024; 131:111911. [PMID: 38527401 DOI: 10.1016/j.intimp.2024.111911] [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: 01/09/2024] [Revised: 02/19/2024] [Accepted: 03/19/2024] [Indexed: 03/27/2024]
Abstract
BACKGROUND Acute lung injury (ALI) has garnered significant attention in the field of respiratory and critical care due to its high mortality and morbidity, and limited treatment options. The role of the endothelial barrier in the development of ALI is crucial. Several bacterial pathogenic factors, including the bacteria-derived formyl peptide (fMLP), have been implicated in damaging the endothelial barrier and initiating ALI. However, the mechanism by which fMLP causes ALI remains unclear. In this study, we aim to explore the mechanisms of ALI caused by fMLP and evaluate the protective effects of MOTS-c, a mitochondrial-derived peptide. METHODS We established a rat model of ALI and a human pulmonary microvascular endothelial cell (HPMVEC) model of ALI by treatment with fMLP. In vivo experiments involved lung histopathology assays, assessments of inflammatory and oxidative stress factors, and measurements of ferroptosis-related proteins and barrier proteins to evaluate the severity of fMLP-induced ALI and the type of tissue damage in rats. In vitro experiments included evaluations of fMLP-induced damage on HPMVEC using cell activity assays, assessments of inflammatory and oxidative stress factors, measurements of ferroptosis-related proteins, endothelial barrier function assays, and examination of the key role of FPR2 in fMLP-induced ALI. We also assessed the protective effect of MOTS-c and investigated its mechanism on the fMLP-induced ALI in vivo and in vitro. RESULTS Results from both in vitro and in vivo experiments demonstrate that fMLP promotes the expression of inflammatory and oxidative stress factors, activates ferroptosis and disrupts the vascular endothelial barrier, ultimately contributing to the development and progression of ALI. Mechanistically, ferroptosis mediated by FPR2 plays a key role in fMLP-induced injury, and the Nrf2 and MAPK pathways are involved in this process. Knockdown of FPR2 and inhibition of ferroptosis can attenuate ALI induced by fMLP. Moreover, MOTS-c could protect the vascular endothelial barrier function by inhibiting ferroptosis and suppressing the expression of inflammatory and oxidative stress factors through Nrf2 and MAPK pathways, thereby alleviating fMLP-induced ALI. CONCLUSION Overall, fMLP disrupts the vascular endothelial barrier through FPR2-mediated ferroptosis, leading to the development and progression of ALI. MOTS-c demonstrates potential as a protective treatment against ALI by alleviating the damage induced by fMLP.
Collapse
Affiliation(s)
- Ziang Wen
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Jidan Fan
- Department of Cardiovascular Surgery, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu 225300, China
| | - Faliang Zhan
- Department of Cardiothoracic Surgery, Yili Friendship Hospital, Yining, Xinjiang Uyghur Autonomous Region 839300, China
| | - Xiaopei Li
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Ben Li
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Peng Lu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Xin Yao
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Zihao Shen
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Zhaoyang Liu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Chufan Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Xiangyu Li
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Wanjun Jin
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Xiao Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Yuanpu Qi
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Xiaowei Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China; Department of Cardiovascular Surgery, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu 225300, China
| | - Meijuan Song
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| |
Collapse
|
29
|
Johnson D, Colijn S, Richee J, Yano J, Burns M, Davis AE, Pham VN, Saric A, Jain A, Yin Y, Castranova D, Melani M, Fujita M, Grainger S, Bonifacino JS, Weinstein BM, Stratman AN. Regulation of angiogenesis by endocytic trafficking mediated by cytoplasmic dynein 1 light intermediate chain 1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.01.587559. [PMID: 38903077 PMCID: PMC11188074 DOI: 10.1101/2024.04.01.587559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
Dynein cytoplasmic 1 light intermediate chain 1 (LIC1, DYNC1LI1) is a core subunit of the dynein motor complex. The LIC1 subunit also interacts with various cargo adaptors to regulate Rab-mediated endosomal recycling and lysosomal degradation. Defects in this gene are predicted to alter dynein motor function, Rab binding capabilities, and cytoplasmic cargo trafficking. Here, we have identified a dync1li1 zebrafish mutant, harboring a premature stop codon at the exon 12/13 splice acceptor site, that displays increased angiogenesis. In vitro, LIC1-deficient human endothelial cells display increases in cell surface levels of the pro-angiogenic receptor VEGFR2, SRC phosphorylation, and Rab11-mediated endosomal recycling. In vivo, endothelial-specific expression of constitutively active Rab11a leads to excessive angiogenesis, similar to the dync1li1 mutants. Increased angiogenesis is also evident in zebrafish harboring mutations in rilpl1/2, the adaptor proteins that promote Rab docking to Lic1 to mediate lysosomal targeting. These findings suggest that LIC1 and the Rab-adaptor proteins RILPL1 and 2 restrict angiogenesis by promoting degradation of VEGFR2-containing recycling endosomes. Disruption of LIC1- and RILPL1/2-mediated lysosomal targeting increases Rab11-mediated recycling endosome activity, promoting excessive SRC signaling and angiogenesis.
Collapse
Affiliation(s)
- Dymonn Johnson
- Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110
| | - Sarah Colijn
- Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110
| | - Jahmiera Richee
- Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110
| | - Joseph Yano
- Section on Vertebrate Organogenesis, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892
- Cell and Molecular Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Margaret Burns
- Section on Vertebrate Organogenesis, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892
| | - Andrew E. Davis
- Section on Vertebrate Organogenesis, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892
| | - Van N. Pham
- Section on Vertebrate Organogenesis, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892
| | - Amra Saric
- Section on Intracellular Protein Trafficking, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892
- Neurosciences and Mental Health Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
| | - Akansha Jain
- Section on Intracellular Protein Trafficking, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892
| | - Ying Yin
- Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110
| | - Daniel Castranova
- Section on Vertebrate Organogenesis, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892
| | - Mariana Melani
- Section on Vertebrate Organogenesis, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892
- Fundación Instituto Leloir, Buenos Aires, Argentina
- Consejo Nacional De Investigaciones Científicas Y Técnicas (CONICET), Buenos Aires, Argentina
- Departamento De Fisiología, Biología Molecular Y Celular, Facultad De Ciencias Exactas Y Naturales, Universidad De Buenos Aires, Buenos Aires, Argentina
| | - Misato Fujita
- Section on Vertebrate Organogenesis, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892
- Kanagawa University, Kanagawa, 221-8686, Japan
| | - Stephanie Grainger
- Department of Cell Biology, Van Andel Institute, Grand Rapids, MI, 49503
| | - Juan S. Bonifacino
- Section on Intracellular Protein Trafficking, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892
| | - Brant M. Weinstein
- Section on Vertebrate Organogenesis, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892
| | - Amber N. Stratman
- Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110
| |
Collapse
|
30
|
Schueler J, Sjöman H, Kriesi C. Sensor extended imaging workflow for creating fit for purpose models in basic and applied cell biology. Commun Biol 2024; 7:170. [PMID: 38341479 PMCID: PMC10858951 DOI: 10.1038/s42003-024-05843-0] [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: 03/21/2023] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
While various engineering disciplines spent years on developing methods and workflows to increase their R&D efficiency, the field of cell biology has seen limited evolution in the fundamental approaches to interact with living cells. Perturbations are mostly of chemical nature, and physiologically relevant contexts and stimuli are left with limited attention, resulting in a solution space constrained within the boundaries of presently manageable perturbations. To predict in the laboratory how a drug will work in a human patient, cell biology must have a closer look at life and strive to mimic the human being in all his complexity. By implementing an iterative process from perturbation to measurement and vice versa, the authors suggest using a sensor-extended imaging workflow to implement product development practices to cell biology, opening a physiologically relevant solution space for the development of truly translational and predictive fit for purpose in vitro cell models.
Collapse
Affiliation(s)
- Julia Schueler
- Charles River Germany GmbH, Am Flughafen 12-14, 79111, Freiburg, Germany.
| | - Heikki Sjöman
- Vitroscope AS, Leirfossvegen 5d, 7037, Trondheim, Norway
| | - Carlo Kriesi
- Vitroscope AS, Leirfossvegen 5d, 7037, Trondheim, Norway
| |
Collapse
|
31
|
Wu X, Cesarovic N, Falk V, Mazza E, Giampietro C. Mechanical factors influence β-catenin localization and barrier properties. Integr Biol (Camb) 2024; 16:zyae013. [PMID: 38952079 DOI: 10.1093/intbio/zyae013] [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: 02/15/2024] [Revised: 06/03/2024] [Accepted: 06/25/2024] [Indexed: 07/03/2024]
Abstract
Mechanical forces are of major importance in regulating vascular homeostasis by influencing endothelial cell behavior and functions. Adherens junctions are critical sites for mechanotransduction in endothelial cells. β-catenin, a component of adherens junctions and the canonical Wnt signaling pathway, plays a role in mechanoactivation. Evidence suggests that β-catenin is involved in flow sensing and responds to tensional forces, impacting junction dynamics. The mechanoregulation of β-catenin signaling is context-dependent, influenced by the type and duration of mechanical loads. In endothelial cells, β-catenin's nuclear translocation and signaling are influenced by shear stress and strain, affecting endothelial permeability. The study investigates how shear stress, strain, and surface topography impact adherens junction dynamics, regulate β-catenin localization, and influence endothelial barrier properties. Insight box Mechanical loads are potent regulators of endothelial functions through not completely elucidated mechanisms. Surface topography, wall shear stress and cyclic wall deformation contribute overlapping mechanical stimuli to which endothelial monolayer respond to adapt and maintain barrier functions. The use of custom developed flow chamber and bioreactor allows quantifying the response of mature human endothelial to well-defined wall shear stress and gradients of strain. Here, the mechanoregulation of β-catenin by substrate topography, wall shear stress, and cyclic stretch is analyzed and linked to the monolayer control of endothelial permeability.
Collapse
Affiliation(s)
- Xi Wu
- ETH Zürich, DMAVT, Experimental Continuum Mechanics, Leonhardstrasse 21, Zurich 8092, Switzerland
| | - Nikola Cesarovic
- Department of Health Sciences and Technology, ETH Zürich, Leopold-Ruzicka-Weg 4, 8093 Zürich, Switzerland
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Augustenburger Platz 1, 13353 Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Volkmar Falk
- Department of Health Sciences and Technology, ETH Zürich, Leopold-Ruzicka-Weg 4, 8093 Zürich, Switzerland
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Augustenburger Platz 1, 13353 Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Edoardo Mazza
- ETH Zürich, DMAVT, Experimental Continuum Mechanics, Leonhardstrasse 21, Zurich 8092, Switzerland
- EMPA, Swiss Federal Laboratories for Materials Science and Technology, Experimental Continuum Mechanics, Überlandstrasse 129, Dübendorf 8600, Switzerland
| | - Costanza Giampietro
- ETH Zürich, DMAVT, Experimental Continuum Mechanics, Leonhardstrasse 21, Zurich 8092, Switzerland
- EMPA, Swiss Federal Laboratories for Materials Science and Technology, Experimental Continuum Mechanics, Überlandstrasse 129, Dübendorf 8600, Switzerland
| |
Collapse
|
32
|
Panagiotides NG, Poledniczek M, Andreas M, Hülsmann M, Kocher AA, Kopp CW, Piechota-Polanczyk A, Weidenhammer A, Pavo N, Wadowski PP. Myocardial Oedema as a Consequence of Viral Infection and Persistence-A Narrative Review with Focus on COVID-19 and Post COVID Sequelae. Viruses 2024; 16:121. [PMID: 38257821 PMCID: PMC10818479 DOI: 10.3390/v16010121] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 01/02/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Microvascular integrity is a critical factor in myocardial fluid homeostasis. The subtle equilibrium between capillary filtration and lymphatic fluid removal is disturbed during pathological processes leading to inflammation, but also in hypoxia or due to alterations in vascular perfusion and coagulability. The degradation of the glycocalyx as the main component of the endothelial filtration barrier as well as pericyte disintegration results in the accumulation of interstitial and intracellular water. Moreover, lymphatic dysfunction evokes an increase in metabolic waste products, cytokines and inflammatory cells in the interstitial space contributing to myocardial oedema formation. This leads to myocardial stiffness and impaired contractility, eventually resulting in cardiomyocyte apoptosis, myocardial remodelling and fibrosis. The following article reviews pathophysiological inflammatory processes leading to myocardial oedema including myocarditis, ischaemia-reperfusion injury and viral infections with a special focus on the pathomechanisms evoked by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. In addition, clinical implications including potential long-term effects due to viral persistence (long COVID), as well as treatment options, are discussed.
Collapse
Affiliation(s)
- Noel G. Panagiotides
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.G.P.); (M.P.); (M.H.); (A.W.); (N.P.)
| | - Michael Poledniczek
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.G.P.); (M.P.); (M.H.); (A.W.); (N.P.)
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria;
| | - Martin Andreas
- Department of Cardiac Surgery, Medical University of Vienna, 1090 Vienna, Austria; (M.A.); (A.A.K.)
| | - Martin Hülsmann
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.G.P.); (M.P.); (M.H.); (A.W.); (N.P.)
| | - Alfred A. Kocher
- Department of Cardiac Surgery, Medical University of Vienna, 1090 Vienna, Austria; (M.A.); (A.A.K.)
| | - Christoph W. Kopp
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria;
| | | | - Annika Weidenhammer
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.G.P.); (M.P.); (M.H.); (A.W.); (N.P.)
| | - Noemi Pavo
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.G.P.); (M.P.); (M.H.); (A.W.); (N.P.)
| | - Patricia P. Wadowski
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria;
| |
Collapse
|
33
|
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
|
34
|
Wang Q, Liang Q, Dou J, Zhou H, Zeng C, Pan H, Shen Y, Li Q, Liu Y, Leong DT, Jiang W, Wang Y. Breaking through the basement membrane barrier to improve nanotherapeutic delivery to tumours. NATURE NANOTECHNOLOGY 2024; 19:95-105. [PMID: 37709950 DOI: 10.1038/s41565-023-01498-w] [Citation(s) in RCA: 53] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 08/03/2023] [Indexed: 09/16/2023]
Abstract
An effective nanotherapeutic transport from the vasculature to the tumour is crucial for cancer treatment with minimal side effects. Here we demonstrate that, in addition to the endothelial barrier, the tumour vascular basement membrane surrounding the endothelium acts as a formidable mechanical barrier that entraps nanoparticles (NPs) in the subendothelial void, forming perivascular NP pools. Breaking through this basement membrane barrier substantially increases NP extravasation. Using inflammation triggered by local hyperthermia, we develop a cooperative immunodriven strategy to overcome the basement membrane barrier that leads to robust tumour killing. Hyperthermia-triggered accumulation and inflammation of platelets attract neutrophils to the NP pools. The subsequent movement of neutrophils through the basement membrane can release the NPs entrapped in the subendothelial void, resulting in increased NP penetration into deeper tumours. We show the necessity of considering the tumour vascular basement membrane barrier when delivering nanotherapeutics. Understanding this barrier will contribute to developing more effective antitumour therapies.
Collapse
Affiliation(s)
- Qin Wang
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Qirui Liang
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jiaxiang Dou
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Han Zhou
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Cici Zeng
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Huimin Pan
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yanqiong Shen
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Quan Li
- Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Yi Liu
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - David Tai Leong
- Department of Chemical and Biomolecular Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore.
| | - Wei Jiang
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Yucai Wang
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China.
| |
Collapse
|
35
|
Alves SAS, Teixeira DE, Peruchetti DB, Silva LS, Brandão LFP, Caruso-Neves C, Pinheiro AAS. Bradykinin produced during Plasmodium falciparum erythrocytic cycle drives monocyte adhesion to human brain microvascular endothelial cells. Brain Res 2024; 1822:148669. [PMID: 37951562 DOI: 10.1016/j.brainres.2023.148669] [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: 07/20/2023] [Revised: 10/30/2023] [Accepted: 11/06/2023] [Indexed: 11/14/2023]
Abstract
Cerebral malaria (CM) pathogenesis is described as a multistep mechanism. In this context, monocytes have been implicated in CM pathogenesis by increasing the sequestration of infected red blood cells to the brain microvasculature. In disease, endothelial activation is followed by reduced monocyte rolling and increased adhesion. Nowadays, an important challenge is to identify potential pro-inflammatory stimuli that can modulate monocytes behavior. Our group have demonstrated that bradykinin (BK), a pro-inflammatory peptide involved in CM, is generated during the erythrocytic cycle of P. falciparum and is detected in culture supernatant (conditioned medium). Herein we investigated the role of BK in the adhesion of monocytes to endothelial cells of blood brain barrier (BBB). To address this issue human monocytic cell line (THP-1) and human brain microvascular endothelial cells (hBMECs) were used. It was observed that 20% conditioned medium from P. falciparum infected erythrocytes (Pf-iRBC sup) increased the adhesion of THP-1 cells to hBMECs. This effect was mediated by BK through the activation of B2 and B1 receptors and involves the increase in ICAM-1 expression in THP-1 cells. Additionally, it was observed that angiotensin-converting enzyme (ACE) inhibitor, captopril, enhanced the effect of both BK and Pf-iRBC sup on THP-1 adhesion. Together these data show that BK, generated during the erythrocytic cycle of P. falciparum, could play an important role in adhesion of monocytes in endothelial cells lining the BBB.
Collapse
Affiliation(s)
- Sarah A S Alves
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Douglas E Teixeira
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Diogo B Peruchetti
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leandro S Silva
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luiz Felipe P Brandão
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Celso Caruso-Neves
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, INCT-Regenera, Conselho Nacional de Desenvolvimento Científico e Tecnológico/MCTIC, Rio de Janeiro, Brazil; Rio de Janeiro Innovation Network in Nanosystems for Health - NanoSAUDE/FAPERJ, Rio de Janeiro, Brazil
| | - Ana Acacia S Pinheiro
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; Rio de Janeiro Innovation Network in Nanosystems for Health - NanoSAUDE/FAPERJ, Rio de Janeiro, Brazil.
| |
Collapse
|
36
|
Karki P, Li Y, Zhang CO, Ke Y, Promnares K, Birukova AA, Eggerman TL, Bocharov AV, Birukov KG. Amphipathic Helical Peptide L37pA Protects against Lung Vascular Endothelial Dysfunction Caused by Truncated Oxidized Phospholipids via Antagonism with CD36 Receptor. Am J Respir Cell Mol Biol 2024; 70:11-25. [PMID: 37725486 PMCID: PMC10768836 DOI: 10.1165/rcmb.2023-0127oc] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 09/12/2023] [Indexed: 09/21/2023] Open
Abstract
The generation of bioactive truncated oxidized phospholipids (Tr-OxPLs) from oxidation of cell-membrane or circulating lipoproteins is a common feature of various pathological states. Scavenger receptor CD36 is involved in lipid transport and acts as a receptor for Tr-OxPLs. Interestingly, Tr-OxPLs and CD36 are involved in endothelial dysfunction-derived acute lung injury, but the precise mechanistic connections remain unexplored. In the present study, we investigated the role of CD36 in mediating pulmonary endothelial cell (EC) dysfunction caused by Tr-OxPLs. Our results demonstrated that the Tr-OxPLs KOdia-PC, Paz-PC, PGPC, PON-PC, POV-PC, and lysophosphocholine caused an acute EC barrier disruption as revealed by measurements of transendothelial electrical resistance and VE-cadherin immunostaining. More importantly, a synthetic amphipathic helical peptide, L37pA, targeting human CD36 strongly attenuated Tr-OxPL-induced EC permeability. L37pA also suppressed Tr-OxPL-induced endothelial inflammatory activation monitored by mRNA expression of inflammatory cytokines/chemokines and adhesion molecules. In addition, L37pA blocked Tr-OxPL-induced NF-κB activation and tyrosine phosphorylation of Src kinase and VE-cadherin. The Src inhibitor SU6656 attenuated KOdia-PC-induced EC permeability and inflammation, but inhibition of the Toll-like receptors (TLRs) TLR1, TLR2, TLR4, and TLR6 had no such protective effects. CD36-knockout mice were more resistant to Tr-OxPL-induced lung injury. Treatment with L37pA was equally effective in ameliorating Tr-OxPL-induced vascular leak and lung inflammation as determined by an Evans blue extravasation assay and total cell and protein content in BAL fluid. Altogether, these results demonstrate an essential role of CD36 in mediating Tr-OxPL-induced EC dysfunction and suggest a strong therapeutic potential of CD36 inhibitory peptides in mitigating lung injury and inflammation.
Collapse
Affiliation(s)
- Pratap Karki
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Yue Li
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Chen-Ou Zhang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Yunbo Ke
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Kamoltip Promnares
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Anna A. Birukova
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Thomas L. Eggerman
- Department of Laboratory Medicine, Clinical Center, and
- National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | | | - Konstantin G. Birukov
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
37
|
Cherubini M, Erickson S, Padmanaban P, Haberkant P, Stein F, Beltran-Sastre V, Haase K. Flow in fetoplacental-like microvessels in vitro enhances perfusion, barrier function, and matrix stability. SCIENCE ADVANCES 2023; 9:eadj8540. [PMID: 38134282 PMCID: PMC10745711 DOI: 10.1126/sciadv.adj8540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 11/22/2023] [Indexed: 12/24/2023]
Abstract
Proper placental vascularization is vital for pregnancy outcomes, but assessing it with animal models and human explants has limitations. We introduce a 3D in vitro model of human placenta terminal villi including fetal mesenchyme and vascular endothelium. By coculturing HUVEC, placental fibroblasts, and pericytes in a macrofluidic chip with a flow reservoir, we generate fully perfusable fetal microvessels. Pressure-driven flow facilitates microvessel growth and remodeling, resulting in early formation of interconnected and lasting placental-like vascular networks. Computational fluid dynamics simulations predict shear forces, which increase microtissue stiffness, decrease diffusivity, and enhance barrier function as shear stress rises. Mass spectrometry analysis reveals enhanced protein expression with flow, including matrix stability regulators, proteins associated with actin dynamics, and cytoskeleton organization. Our model provides a powerful tool for deducing complex in vivo parameters, such as shear stress on developing vascularized placental tissue, and holds promise for unraveling gestational disorders related to the vasculature.
Collapse
Affiliation(s)
- Marta Cherubini
- European Molecular Biology Laboratory (EMBL), Barcelona, Spain
| | - Scott Erickson
- European Molecular Biology Laboratory (EMBL), Barcelona, Spain
| | | | - Per Haberkant
- Proteomics Core Facility, EMBL Heidelberg, Heidelberg, Germany
| | - Frank Stein
- Proteomics Core Facility, EMBL Heidelberg, Heidelberg, Germany
| | | | - Kristina Haase
- European Molecular Biology Laboratory (EMBL), Barcelona, Spain
| |
Collapse
|
38
|
Zhang Q, Du G, Tong L, Guo X, Wei Y. Overexpression of LOX-1 in hepatocytes protects vascular smooth muscle cells from phenotype transformation and wire injury induced carotid neoatherosclerosis through ALOX15. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166805. [PMID: 37468019 DOI: 10.1016/j.bbadis.2023.166805] [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: 10/30/2022] [Revised: 06/16/2023] [Accepted: 07/06/2023] [Indexed: 07/21/2023]
Abstract
Neoatherosclerosis (NA), the main pathological basis of late stent failure, is the main limitation of interventional therapy. However, the specific pathogenesis and treatment remain unclear. In vivo, NA model was established by carotid wire injury and high-fat feeding in ApoE-/- mice. Oxidized low-density lipoprotein receptor-1/lectin-like oxidized low-density lipoprotein receptor-1 (OLR1/LOX-1), a specific receptor for oxidized low-density lipoprotein (ox-LDL), was specifically ectopically overexpressed in hepatocytes by portal vein injection of adeno-associated serotype 8 (AAV8)-thyroid binding globulin (TBG)-Olr1 and the protective effect against NA was examined. In vitro, LOX-1 was overexpressed on HHL5 using lentivirus (LV)-OLR1 and the vascular smooth muscle cells (VSMCs)-HHL5 indirect co-culture system was established to examine its protective effect on VSMCs and the molecular mechanism. Functionally, we found that specific ectopic overexpression of LOX-1 by hepatocytes competitively engulfed and metabolized ox-LDL, alleviating its resulting phenotypic transformation of VSMCs including migration, downregulation of contractile shape markers (smooth muscle α-actin (SMαA) and smooth muscle-22α (SM22α)), and upregulation of proliferative/migratory shape markers (osteopontin (OPN) and Vimentin) as well as foaminess and apoptosis, thereby alleviating NA, which independent of low-density lipoprotein (LDL) lowering treatment (evolocumab, a monoclonal antibody to proprotein convertase subtilisin/kexin type 9 (PCSK9)). Mechanistically, we found that overexpression of LOX-1 in hepatocytes competitively engulfed and metabolized ox-LDL through upregulation of arachidonate-15-lipoxygenase (ALOX15), which further upregulated scavenger receptor class B type I (SRBI) and ATP-binding cassette transporter A1 (ABCA1). In conclusion, the overexpression of LOX-1 in liver protects VSMCs from phenotypic transformation and wire injury induced carotid neoatherosclerosis through ALOX15.
Collapse
Affiliation(s)
- Qing Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gaohui Du
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lu Tong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaopeng Guo
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yumiao Wei
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
39
|
You Q, Wu J, Liu Y, Zhang F, Jiang N, Tian X, Cai Y, Yang E, Lyu R, Zheng N, Chen D, Wu Z. HMGB1 Release Induced by EV71 Infection Exacerbates Blood-Brain Barrier Disruption via VE-cadherin Phosphorylation. Virus Res 2023; 338:199240. [PMID: 37832655 PMCID: PMC10587765 DOI: 10.1016/j.virusres.2023.199240] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 10/15/2023]
Abstract
PURPOSE EV71 (Enterovirus 71) is a major causative agent of the outbreaks of HFMD (hand, foot, and mouth disease), which is associated with neurological damage caused by permeability disruption of BBB (blood-brain barrier). HMGB1 (high-mobility group box 1) is a widely expressed nuclear protein that triggers host inflammatory responses. Our work aimed to explore the function of HMGB1 in EV71 infection and its contributions to EV71-related BBB damage. METHODS HeLa cells, HT-29 cells and AG6 mice were used to explore the translocation of HMGB1 in EV71 infection in vitro and in vivo. The roles of released HMGB1 on EV71 replication and associated inflammatory cytokines were investigated using recombinant HMGB1 in HeLa cells. The mechanisms of released HMGB1 in EV71-induced BBB injury were explored using recombinant HMGB1 and anti-HMGB1 neutralizing antibodies in monolayer HCMECs (immortalized human brain microvascular endothelial cells) and AG6 mice brain. RESULTS EV71 induced HMGB1 nucleocytoplasmic translocation and extracellular release in vitro and in vivo. Released HMGB1 acted as an inflammatory mediator in EV71 infection rather than affecting viral replication in vitro. Released HMGB1 disrupted BBB integrity by enhancing VE-cadherin phosphorylation at tyrosine 685 in HCMECs, and reducing total VE-cadherin levels in HCMECs and AG6 mice in EV71 infection. And released HMGB1 induced an increase in activated astrocytes. Neutralization of HMGB1 reversed the increased endothelial hyperpermeability and phosphorylation of VE-cadherin in HCMECs. CONCLUSION The inflammatory mediator HMGB1 released by EV71 exacerbated BBB disruption by enhancing VE-cadherin phosphorylation, which in turn aggravated EV71-induced neuroinflammation.
Collapse
Affiliation(s)
- Qiao You
- Center for Public Health Research, Medical School of Nanjing University, Nanjing, China
| | - Jing Wu
- Center for Public Health Research, Medical School of Nanjing University, Nanjing, China
| | - Ye Liu
- China Department of Ophthalmology, Tianjin First Central Hospital, Tianjin, China
| | - Fang Zhang
- Department of Burn and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Na Jiang
- Center for Public Health Research, Medical School of Nanjing University, Nanjing, China
| | - Xiaoyan Tian
- Center for Public Health Research, Medical School of Nanjing University, Nanjing, China
| | - Yurong Cai
- School of Life Sciences, Ningxia University, Yinchuan, PR China
| | - Enhui Yang
- Department of Child Healthcare, Wenzhou People's Hospital, Wenzhou, Zhejiang, China
| | - Ruining Lyu
- Center for Public Health Research, Medical School of Nanjing University, Nanjing, China
| | - Nan Zheng
- Center for Public Health Research, Medical School of Nanjing University, Nanjing, China
| | - Deyan Chen
- Center for Public Health Research, Medical School of Nanjing University, Nanjing, China.
| | - Zhiwei Wu
- School of Life Sciences, Ningxia University, Yinchuan, PR China; State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China; Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China.
| |
Collapse
|
40
|
Shadab M, Slavin SA, Mahamed Z, Millar MW, Najar RA, Leonard A, Pietropaoli A, Dean DA, Fazal F, Rahman A. Spleen Tyrosine Kinase phosphorylates VE-cadherin to cause endothelial barrier disruption in acute lung injury. J Biol Chem 2023; 299:105408. [PMID: 38229397 PMCID: PMC10731244 DOI: 10.1016/j.jbc.2023.105408] [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: 06/08/2023] [Revised: 09/24/2023] [Accepted: 10/10/2023] [Indexed: 01/18/2024] Open
Abstract
Increased endothelial cell (EC) permeability is a cardinal feature of acute lung injury/acute respiratory distress syndrome (ALI/ARDS). Tyrosine phosphorylation of VE-cadherin is a key determinant of EC barrier disruption. However, the identity and role of tyrosine kinases in this context are incompletely understood. Here we report that Spleen Tyrosine Kinase (Syk) is a key mediator of EC barrier disruption and lung vascular leak in sepsis. Inhibition of Syk by pharmacological or genetic approaches, each reduced thrombin-induced EC permeability. Mechanistically, Syk associates with and phosphorylates VE-cadherin to cause EC permeability. To study the causal role of endothelial Syk in sepsis-induced ALI, we used a remarkably efficient and cost-effective approach based on gene transfer to generate EC-ablated Syk mice. These mice were protected against sepsis-induced loss of VE-cadherin and inflammatory lung injury. Notably, the administration of Syk inhibitor R788 (fostamatinib); currently in phase II clinical trial for the treatment of COVID-19, mitigated lung injury and mortality in mice with sepsis. These data identify Syk as a novel kinase for VE-cadherin and a druggable target against ALI in sepsis.
Collapse
Affiliation(s)
- Mohammad Shadab
- Department of Pediatrics, Lung Biology and Disease Program, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Spencer A Slavin
- Department of Pediatrics, Lung Biology and Disease Program, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Zahra Mahamed
- Department of Pediatrics, Lung Biology and Disease Program, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Michelle W Millar
- Department of Pediatrics, Lung Biology and Disease Program, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Rauf A Najar
- Department of Pediatrics, Lung Biology and Disease Program, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Antony Leonard
- Department of Pediatrics, Lung Biology and Disease Program, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Anthony Pietropaoli
- Department of Medicine, Lung Biology and Disease Program, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - David A Dean
- Department of Pediatrics, Lung Biology and Disease Program, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Fabeha Fazal
- Department of Pediatrics, Lung Biology and Disease Program, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Arshad Rahman
- Department of Pediatrics, Lung Biology and Disease Program, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA.
| |
Collapse
|
41
|
Tang J, Kang Y, Zhou Y, Shang N, Li X, Wang H, Lan J, Wang S, Wu L, Peng Y. TIMP2 ameliorates blood-brain barrier disruption in traumatic brain injury by inhibiting Src-dependent VE-cadherin internalization. J Clin Invest 2023; 134:e164199. [PMID: 38015626 PMCID: PMC10849766 DOI: 10.1172/jci164199] [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: 08/05/2022] [Accepted: 11/21/2023] [Indexed: 11/30/2023] Open
Abstract
Blood-brain barrier (BBB) disruption is a serious pathological consequence of traumatic brain injury (TBI), for which there are limited therapeutic strategies. Tissue inhibitor of metalloproteinase-2 (TIMP2), a molecule with dual functions of inhibiting MMP activity and displaying cytokine-like activity through receptor binding, has been reported to inhibit VEGF-induced vascular hyperpermeability. Here, we investigate the ability of TIMP2 to ameliorate BBB disruption in TBI and the underlying molecular mechanisms. Both TIMP2 and AlaTIMP2, a TIMP2 mutant without MMP-inhibiting activity, attenuated neurological deficits and BBB leakage in TBI mice; they also inhibited junctional protein degradation and translocation to reduce paracellular permeability in human brain microvascular endothelial cells (ECs) exposed to hypoxic plus inflammatory insult. Mechanistic studies revealed that TIMP2 interacted with α3β1 integrin on ECs, inhibiting Src activation-dependent VE-cadherin phosphorylation, VE-cadherin/catenin complex destabilization, and subsequent VE-cadherin internalization. Notably, localization of VE-cadherin on the membrane was critical for TIMP2-mediated EC barrier integrity. Furthermore, TIMP2-mediated increased membrane localization of VE-cadherin enhanced the level of active Rac1, thereby inhibiting stress fiber formation. All together, our studies have identified an MMP-independent mechanism by which TIMP2 regulates EC barrier integrity after TBI. TIMP2 may be a therapeutic agent for TBI and other neurological disorders involving BBB breakdown.
Collapse
|
42
|
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
|
43
|
Tamargo IA, Baek KI, Kim Y, Park C, Jo H. Flow-induced reprogramming of endothelial cells in atherosclerosis. Nat Rev Cardiol 2023; 20:738-753. [PMID: 37225873 PMCID: PMC10206587 DOI: 10.1038/s41569-023-00883-1] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/25/2023] [Indexed: 05/26/2023]
Abstract
Atherosclerotic diseases such as myocardial infarction, ischaemic stroke and peripheral artery disease continue to be leading causes of death worldwide despite the success of treatments with cholesterol-lowering drugs and drug-eluting stents, raising the need to identify additional therapeutic targets. Interestingly, atherosclerosis preferentially develops in curved and branching arterial regions, where endothelial cells are exposed to disturbed blood flow with characteristic low-magnitude oscillatory shear stress. By contrast, straight arterial regions exposed to stable flow, which is associated with high-magnitude, unidirectional shear stress, are relatively well protected from the disease through shear-dependent, atheroprotective endothelial cell responses. Flow potently regulates structural, functional, transcriptomic, epigenomic and metabolic changes in endothelial cells through mechanosensors and mechanosignal transduction pathways. A study using single-cell RNA sequencing and chromatin accessibility analysis in a mouse model of flow-induced atherosclerosis demonstrated that disturbed flow reprogrammes arterial endothelial cells in situ from healthy phenotypes to diseased ones characterized by endothelial inflammation, endothelial-to-mesenchymal transition, endothelial-to-immune cell-like transition and metabolic changes. In this Review, we discuss this emerging concept of disturbed-flow-induced reprogramming of endothelial cells (FIRE) as a potential pro-atherogenic mechanism. Defining the flow-induced mechanisms through which endothelial cells are reprogrammed to promote atherosclerosis is a crucial area of research that could lead to the identification of novel therapeutic targets to combat the high prevalence of atherosclerotic disease.
Collapse
Affiliation(s)
- Ian A Tamargo
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
- Molecular and Systems Pharmacology Program, Emory University, Atlanta, GA, USA
| | - Kyung In Baek
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Yerin Kim
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Christian Park
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA.
- Molecular and Systems Pharmacology Program, Emory University, Atlanta, GA, USA.
- Department of Medicine, Emory University School, Atlanta, GA, USA.
| |
Collapse
|
44
|
Ryø LB, Haslund D, Rovsing AB, Pihl R, Sanrattana W, de Maat S, Palarasah Y, Maas C, Thiel S, Mikkelsen JG. Restriction of C1-inhibitor activity in hereditary angioedema by dominant-negative effects of disease-associated SERPING1 gene variants. J Allergy Clin Immunol 2023; 152:1218-1236.e9. [PMID: 37301409 DOI: 10.1016/j.jaci.2023.04.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 04/17/2023] [Accepted: 04/25/2023] [Indexed: 06/12/2023]
Abstract
BACKGROUND Patients with hereditary angioedema experience recurrent, sometimes life-threatening, attacks of edema. It is a rare genetic disorder characterized by genetic and clinical heterogenicity. Most cases are caused by genetic variants in the SERPING1 gene leading to plasma deficiency of the encoded protein C1 inhibitor (C1INH). More than 500 different hereditary angioedema-causing variants have been identified in the SERPING1 gene, but the disease mechanisms by which they result in pathologically low C1INH plasma levels remain largely unknown. OBJECTIVES The aim was to describe trans-inhibitory effects of full-length or near full-length C1INH encoded by 28 disease-associated SERPING1 variants. METHODS HeLa cells were transfected with expression constructs encoding the studied SERPING1 variants. Extensive and comparative studies of C1INH expression, secretion, functionality, and intracellular localization were carried out. RESULTS Our findings characterized functional properties of a subset of SERPING1 variants allowing the examined variants to be subdivided into 5 different clusters, each containing variants sharing specific molecular characteristics. For all variants except 2, we found that coexpression of mutant and normal C1INH negatively affected the overall capacity to target proteases. Strikingly, for a subset of variants, intracellular formation of C1INH foci was detectable only in heterozygous configurations enabling simultaneous expression of normal and mutant C1INH. CONCLUSIONS We provide a functional classification of SERPING1 gene variants suggesting that different SERPING1 variants drive the pathogenicity through different and in some cases overlapping molecular disease mechanisms. For a subset of gene variants, our data define some types of hereditary angioedema with C1INH deficiency as serpinopathies driven by dominant-negative disease mechanisms.
Collapse
Affiliation(s)
| | - Didde Haslund
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Rasmus Pihl
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Wariya Sanrattana
- CDL Research, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Steven de Maat
- CDL Research, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Yaseelan Palarasah
- Department of Cancer and Inflammation Research, University of Southern Denmark, Odense, Denmark; Department of Clinical Biochemistry, Hospital of South West Jutland, Esbjerg, Denmark
| | - Coen Maas
- CDL Research, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands; Unit for Thrombosis Research, Department of Regional Health Research, University of Southern Denmark, Esbjerg, Denmark
| | - Steffen Thiel
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | |
Collapse
|
45
|
Tiruppathi C, Wang DM, Ansari MO, Bano S, Tsukasaki Y, Mukhopadhyay A, Joshi JC, Loch C, Niessen HWM, Malik AB. Ubiquitin ligase CHFR mediated degradation of VE-cadherin through ubiquitylation disrupts endothelial adherens junctions. Nat Commun 2023; 14:6582. [PMID: 37852964 PMCID: PMC10584835 DOI: 10.1038/s41467-023-42225-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 10/04/2023] [Indexed: 10/20/2023] Open
Abstract
Vascular endothelial cadherin (VE-cadherin) expressed at endothelial adherens junctions (AJs) is vital for vascular integrity and endothelial homeostasis. Here we identify the requirement of the ubiquitin E3-ligase CHFR as a key mechanism of ubiquitylation-dependent degradation of VE-cadherin. CHFR was essential for disrupting the endothelium through control of the VE-cadherin protein expression at AJs. We observe augmented expression of VE-cadherin in endothelial cell (EC)-restricted Chfr knockout (ChfrΔEC) mice. We also observe abrogation of LPS-induced degradation of VE-cadherin in ChfrΔEC mice, suggesting the pathophysiological relevance of CHFR in regulating the endothelial junctional barrier in inflammation. Lung endothelial barrier breakdown, inflammatory neutrophil extravasation, and mortality induced by LPS were all suppressed in ChfrΔEC mice. We find that the transcription factor FoxO1 is a key upstream regulator of CHFR expression. These findings demonstrate the requisite role of the endothelial cell-expressed E3-ligase CHFR in regulating the expression of VE-cadherin, and thereby endothelial junctional barrier integrity.
Collapse
Affiliation(s)
- Chinnaswamy Tiruppathi
- Department of Pharmacology and Regenerative Medicine and The Center of Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL, USA.
| | - Dong-Mei Wang
- Department of Pharmacology and Regenerative Medicine and The Center of Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL, USA
| | - Mohammad Owais Ansari
- Department of Pharmacology and Regenerative Medicine and The Center of Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL, USA
| | - Shabana Bano
- Department of Pharmacology and Regenerative Medicine and The Center of Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL, USA
| | - Yoshikazu Tsukasaki
- Department of Pharmacology and Regenerative Medicine and The Center of Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL, USA
| | - Amitabha Mukhopadhyay
- Department of Pharmacology and Regenerative Medicine and The Center of Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL, USA
| | - Jagdish C Joshi
- Department of Pharmacology and Regenerative Medicine and The Center of Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL, USA
| | | | - Hans W M Niessen
- Department of Pathology, VU University Medical Center, Amsterdam, the Netherlands
| | - Asrar B Malik
- Department of Pharmacology and Regenerative Medicine and The Center of Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL, USA.
| |
Collapse
|
46
|
Sjöberg E, Melssen M, Richards M, Ding Y, Chanoca C, Chen D, Nwadozi E, Pal S, Love DT, Ninchoji T, Shibuya M, Simons M, Dimberg A, Claesson-Welsh L. Endothelial VEGFR2-PLCγ signaling regulates vascular permeability and antitumor immunity through eNOS/Src. J Clin Invest 2023; 133:e161366. [PMID: 37651195 PMCID: PMC10575733 DOI: 10.1172/jci161366] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 08/15/2023] [Indexed: 09/02/2023] Open
Abstract
Endothelial phospholipase Cγ (PLCγ) is essential for vascular development; however, its role in healthy, mature, or pathological vessels is unexplored. Here, we show that PLCγ was prominently expressed in vessels of several human cancer forms, notably in renal cell carcinoma (RCC). High PLCγ expression in clear cell RCC correlated with angiogenic activity and poor prognosis, while low expression correlated with immune cell activation. PLCγ was induced downstream of vascular endothelial growth factor receptor 2 (VEGFR2) phosphosite Y1173 (pY1173). Heterozygous Vegfr2Y1173F/+ mice or mice lacking endothelial PLCγ (Plcg1iECKO) exhibited a stabilized endothelial barrier and diminished vascular leakage. Barrier stabilization was accompanied by decreased expression of immunosuppressive cytokines, reduced infiltration of B cells, helper T cells and regulatory T cells, and improved response to chemo- and immunotherapy. Mechanistically, pY1173/PLCγ signaling induced Ca2+/protein kinase C-dependent activation of endothelial nitric oxide synthase (eNOS), required for tyrosine nitration and activation of Src. Src-induced phosphorylation of VE-cadherin at Y685 was accompanied by disintegration of endothelial junctions. This pY1173/PLCγ/eNOS/Src pathway was detected in both healthy and tumor vessels in Vegfr2Y1173F/+ mice, which displayed decreased activation of PLCγ and eNOS and suppressed vascular leakage. Thus, we believe that we have identified a clinically relevant endothelial PLCγ pathway downstream of VEGFR2 pY1173, which destabilizes the endothelial barrier and results in loss of antitumor immunity.
Collapse
Affiliation(s)
- Elin Sjöberg
- Department of Immunology, Genetics and Pathology, Beijer and Science for Life Laboratories, Uppsala University, Uppsala, Sweden
| | - Marit Melssen
- Department of Immunology, Genetics and Pathology, Beijer and Science for Life Laboratories, Uppsala University, Uppsala, Sweden
| | - Mark Richards
- Department of Immunology, Genetics and Pathology, Beijer and Science for Life Laboratories, Uppsala University, Uppsala, Sweden
| | - Yindi Ding
- Department of Immunology, Genetics and Pathology, Beijer and Science for Life Laboratories, Uppsala University, Uppsala, Sweden
| | - Catarina Chanoca
- Department of Immunology, Genetics and Pathology, Beijer and Science for Life Laboratories, Uppsala University, Uppsala, Sweden
| | - Dongying Chen
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Emmanuel Nwadozi
- Department of Immunology, Genetics and Pathology, Beijer and Science for Life Laboratories, Uppsala University, Uppsala, Sweden
| | - Sagnik Pal
- Department of Immunology, Genetics and Pathology, Beijer and Science for Life Laboratories, Uppsala University, Uppsala, Sweden
| | - Dominic T. Love
- Department of Immunology, Genetics and Pathology, Beijer and Science for Life Laboratories, Uppsala University, Uppsala, Sweden
| | - Takeshi Ninchoji
- Department of Immunology, Genetics and Pathology, Beijer and Science for Life Laboratories, Uppsala University, Uppsala, Sweden
| | - Masabumi Shibuya
- Institute of Physiology and Medicine, Jobu University, Takasaki, Gunma, Japan
| | - Michael Simons
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Anna Dimberg
- Department of Immunology, Genetics and Pathology, Beijer and Science for Life Laboratories, Uppsala University, Uppsala, Sweden
| | - Lena Claesson-Welsh
- Department of Immunology, Genetics and Pathology, Beijer and Science for Life Laboratories, Uppsala University, Uppsala, Sweden
| |
Collapse
|
47
|
McQueen A, Warboys CM. Mechanosignalling pathways that regulate endothelial barrier function. Curr Opin Cell Biol 2023; 84:102213. [PMID: 37531894 DOI: 10.1016/j.ceb.2023.102213] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/27/2023] [Accepted: 07/04/2023] [Indexed: 08/04/2023]
Abstract
Blood vessels are lined by a single layer of endothelial cells that provide a barrier between circulating plasma and the underlying tissue. Permeability of endothelial cells is tightly regulated, and increased permeability is associated with a number of diseases including atherosclerosis. Endothelial cells are continuously exposed to mechanical forces exerted by flowing blood and are particularly sensitive to shear stress, which is a key determinant of endothelial function. Undisturbed flow promotes endothelial resilience and reduces permeability to macromolecules whereas disturbed flow promotes endothelial dysfunction and barrier disruption. This review will outline recent advances in our understanding of how disturbed and undisturbed flow regulate paracellular and transcellular permeability and will highlight potential cellular targets that could form the basis of therapies to limit the development of cardiovascular disease.
Collapse
Affiliation(s)
- Anna McQueen
- Department of Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, London, NW1 0NN, UK
| | - Christina M Warboys
- Department of Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, London, NW1 0NN, UK.
| |
Collapse
|
48
|
Breslin JW. Edema and lymphatic clearance: molecular mechanisms and ongoing challenges. Clin Sci (Lond) 2023; 137:1451-1476. [PMID: 37732545 PMCID: PMC11025659 DOI: 10.1042/cs20220314] [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: 06/03/2023] [Revised: 08/18/2023] [Accepted: 08/31/2023] [Indexed: 09/22/2023]
Abstract
Resolution of edema remains a significant clinical challenge. Conditions such as traumatic shock, sepsis, or diabetes often involve microvascular hyperpermeability, which leads to tissue and organ dysfunction. Lymphatic insufficiency due to genetic causes, surgical removal of lymph nodes, or infections, leads to varying degrees of tissue swelling that impair mobility and immune defenses. Treatment options are limited to management of edema as there are no specific therapeutics that have demonstrated significant success for ameliorating microvascular leakage or impaired lymphatic function. This review examines current knowledge about the physiological, cellular, and molecular mechanisms that control microvascular permeability and lymphatic clearance, the respective processes for interstitial fluid formation and removal. Clinical conditions featuring edema, along with potential future directions are discussed.
Collapse
Affiliation(s)
- Jerome W Breslin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, FL, U.S.A
| |
Collapse
|
49
|
Chovatiya G, Li KN, Li J, Ghuwalewala S, Tumbar T. Alk1 acts in non-endothelial VE-cadherin + perineurial cells to maintain nerve branching during hair homeostasis. Nat Commun 2023; 14:5623. [PMID: 37699906 PMCID: PMC10497554 DOI: 10.1038/s41467-023-40761-5] [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: 08/02/2022] [Accepted: 08/09/2023] [Indexed: 09/14/2023] Open
Abstract
Vascular endothelial (VE)-cadherin is a well-recognized endothelial cell marker. One of its interacting partners, the TGF-β receptor Alk1, is essential in endothelial cells for adult skin vasculature remodeling during hair homeostasis. Using single-cell transcriptomics, lineage tracing and gene targeting in mice, we characterize the cellular and molecular dynamics of skin VE-cadherin+ cells during hair homeostasis. We describe dynamic changes of VE-cadherin+ endothelial cells specific to blood and lymphatic vessels and uncover an atypical VE-cadherin+ cell population. The latter is not a predicted adult endovascular progenitor, but rather a non-endothelial mesenchymal perineurial cell type, which forms nerve encapsulating tubular structures that undergo remodeling during hair homeostasis. Alk1 acts in the VE-cadherin+ perineurial cells to maintain proper homeostatic nerve branching by enforcing basement membrane and extracellular matrix molecular signatures. Our work implicates the VE-cadherin/Alk1 duo, classically known as endothelial-vascular specific, in perineurial-nerve homeostasis. This has broad implications in vascular and nerve disease.
Collapse
Affiliation(s)
- Gopal Chovatiya
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Kefei Nina Li
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Jonathan Li
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Sangeeta Ghuwalewala
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Tudorita Tumbar
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
50
|
Kizhatil K, Clark G, Sunderland D, Bhandari A, Horbal L, Balasubramanian R, John S. FYN regulates aqueous humor outflow and IOP through the phosphorylation of VE-cadherin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.04.556253. [PMID: 37886565 PMCID: PMC10602025 DOI: 10.1101/2023.09.04.556253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
The exact sites and molecules that determine resistance to aqueous humor drainage and control intraocular pressure (IOP) need further elaboration. Proposed sites include the inner wall of Schlemms's canal and the juxtacanalicular trabecular meshwork ocular drainage tissues. The adherens junctions (AJs) of Schlemm's canal endothelial cells (SECs) must both preserve the blood-aqueous humor (AQH) barrier and be conducive to AQH drainage. How homeostatic control of AJ permeability in SC occurs and how such control impacts IOP is unclear. We hypothesized that mechano-responsive phosphorylation of the junctional molecule VE-CADHERIN (VEC) by SRC family kinases (SFKs) regulates the permeability of SEC AJs. We tested this by clamping IOP at either 16 mmHg, 25 mmHg, or 45 mmHg in mice and then measuring AJ permeability and VEC phosphorylation. We found that with increasing IOP: 1) SEC AJ permeability increased, 2) VEC phosphorylation was increased at tyrosine-658, and 3) SFKs were activated at the AJ. Among the two SFKs known to phosphorylate VEC, FYN, but not SRC, localizes to the SC. Furthermore, FYN mutant mice had decreased phosphorylation of VEC at SEC AJs, dysregulated IOP, and reduced AQH outflow. Together, our data demonstrate that increased IOP activates FYN in the inner wall of SC, leading to increased phosphorylation of AJ VEC and, thus, decreased resistance to AQH outflow. These findings support a crucial role of mechanotransduction signaling in IOP homeostasis within SC in response to IOP. These data strongly suggest that the inner wall of SC partially contributes to outflow resistance.
Collapse
|