1
|
Pan Z, Yao Q, Kong W, Ma X, Tian L, Zhao Y, Zhu S, Chen S, Sun M, Liu J, Jiang S, Ma J, Liu Q, Peng X, Li X, Hong Z, Hong Y, Wang X, Liu J, Zhang J, Zhang W, Sun B, Pahlavan S, Xia Y, Shen W, Liu Y, Jiang W, Xie Z, Kong W, Wang X, Wang K. Generation of iPSC-derived human venous endothelial cells for the modeling of vascular malformations and drug discovery. Cell Stem Cell 2025; 32:227-245.e9. [PMID: 39579761 DOI: 10.1016/j.stem.2024.10.015] [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: 05/13/2024] [Revised: 09/04/2024] [Accepted: 10/28/2024] [Indexed: 11/25/2024]
Abstract
Venous malformations (VMs) represent prevalent vascular anomalies typically attributed to non-inherited somatic mutations within venous endothelial cells (VECs). The lack of robust disease models for VMs impedes drug discovery. Here, we devise a robust protocol for the generation of human induced VECs (iVECs) through manipulation of cell-cycle dynamics via the retinoic signaling pathway. We introduce an L914F mutation into the TIE2 gene locus of induced pluripotent stem cells (iPSCs) and show that the mutated iVECs form dilated blood vessels after transplantation into mice, thereby recapitulating the phenotypic characteristics observed in VMs. Moreover, utilizing a deep neural network and a high-throughput digital RNA with perturbation of genes sequencing (DRUG-seq) approach, we perform drug screening and demonstrate that bosutinib effectively rescues the disease phenotype in vitro and in vivo. In summary, by leveraging genome editing and stem cell technology, we generate VM models that enable the development of additional therapeutics.
Collapse
Affiliation(s)
- Zihang Pan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Qiyang Yao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Weijing Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Xiaojing Ma
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Liangliang Tian
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Yun Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Shuntian Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Sheng Chen
- Department of Burns and Plastic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Mengze Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Jiao Liu
- Center of Medical and Health Analysis, Peking University Health Science Center, Beijing 100191, China
| | - Simin Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Jianxun Ma
- Department of Plastic Surgery, Peking University Third Hospital, Beijing 100191, China
| | - Qijia Liu
- Department of Interventional Radiology and Vascular Surgery, Peking University Third Hospital, Beijing 100191, China
| | - Xiaohong Peng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Xiaoxia Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Zixuan Hong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Yi Hong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Xue Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Jiarui Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Jingjing Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Wei Zhang
- TianXinFu (Beijing) Medical Appliance Co., Ltd., Beijing 102200, China
| | - Bingbing Sun
- TianXinFu (Beijing) Medical Appliance Co., Ltd., Beijing 102200, China
| | - Sara Pahlavan
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Youchen Xia
- Department of Plastic Surgery, Peking University Third Hospital, Beijing 100191, China
| | - Weimin Shen
- Department of Burns and Plastic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuyong Liu
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Wenjian Jiang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Zhengwei Xie
- Peking University International Cancer Institute, Health Science Center, Peking University, Beijing 100191, China.
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China.
| | - Xi Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China; State Key Laboratory of Female Fertility Promotion, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China; Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, China.
| | - Kai Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China.
| |
Collapse
|
2
|
Rustagi V, Rameshwari R, Kumar Singh I. Identification of potential inhibitors for MAP4K4 in glaucoma using meta-dynamics-based dissociation free energy calculation. Brain Res 2025; 1847:149300. [PMID: 39500479 DOI: 10.1016/j.brainres.2024.149300] [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: 05/23/2024] [Revised: 10/16/2024] [Accepted: 10/26/2024] [Indexed: 11/12/2024]
Abstract
Glaucoma, a prevalent eye ailment causing irreversible vision loss, affects over 295 million individuals globally, necessitating the exploration of novel therapeutic avenues. Despite extensive research on targets like the phosphodiesterase enzyme and rho kinase, the potential of MAP4K4 in glaucoma remains untapped. This study aims to identify potent MAP4K4 inhibitors to counteract retinal cell apoptosis and oxidative stress associated with glaucoma. Using HTVS and XP docking, 911,059 compounds were screened. The MMGBSA calculation and pharmacokinetics analysis were used to shortlist the compounds. After performing 75 molecular dynamics simulations, further meta-dynamics were employed to calculate dissociation-free energy and find potential MAP4K4 inhibitors. Findings indicated that ZINC06717217 and ZINC38836256 exhibited remarkable promise, with docking scores of -9.57 and -11.12 and MMGBSA binding energies of -91.07 kcal/mol and -87.52 kcal/mol, respectively. Comparative analysis with the reference compound Q27453723 underscored their superior stability, requiring dissociation-free energies of -15.11 kcal/mol and -12.46 kcal/mol to disengage from the docked complex. This underscored their robust binding affinity. ZINC06717217 and ZINC38836256 show promising stability and strong binding to the MAP4K4 protein. Hence, these findings are promising in inhibiting MAP4K4 for glaucoma treatment, potentially leading to more effective treatment and curing blindness. KEY MESSAGES: First to incorporate the dissociation-free energy for identifying compounds for glaucoma treatment. In-silico analysis showed that ZINC06717217 and ZINC38836256 are promising compounds for targeting MAP4K4.
Collapse
Affiliation(s)
- Vanshika Rustagi
- Department of Biotechnology, School of Engineering and Technology, Manav Rachna International Institute of Research and Studies, Faridabad, Haryana 121004, India
| | - Rashmi Rameshwari
- Department of Biotechnology, School of Engineering and Technology, Manav Rachna International Institute of Research and Studies, Faridabad, Haryana 121004, India.
| | - Indrakant Kumar Singh
- Molecular Biology Research Lab, Department of Zoology, Deshbandhu College, University of Delhi, Kalkaji, New Delhi 110019, India; Delhi School of Public Health, Institute of Eminence, University of Delhi, Delhi 110007, India.
| |
Collapse
|
3
|
Joachim J, Maselli D, Petsolari E, Aman J, Swiatlowska P, Killock D, Chaudhry H, Zarban AA, Sarker M, Fraser P, Cleary SJ, Amison R, Cuthbert I, Yang Y, Meier M, Fraternali F, Brain SD, Shah AM, Ivetic A. TNIK: A redox sensor in endothelial cell permeability. SCIENCE ADVANCES 2024; 10:eadk6583. [PMID: 39705357 DOI: 10.1126/sciadv.adk6583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 11/18/2024] [Indexed: 12/22/2024]
Abstract
Dysregulation of endothelial barrier integrity can lead to vascular leak and potentially fatal oedema. TNF-α controls endothelial permeability during inflammation and requires the actin organizing Ezrin-Radixin-Moesin (ERM) proteins. We identified TRAF2 and NCK-interacting kinase (TNIK) as a kinase directly phosphorylating and activating ERM, specifically at the plasma membrane of primary human endothelial cells. TNIK mediates TNF-α-dependent cellular stiffness and paracellular gap formation in vitro and is essential in driving inflammatory oedema formation in vivo. Unlike its homologs, TNIK activity is negatively and reversibly regulated by H2O2-mediated oxidation of C202 within the kinase domain. TNIK oxidation results in intermolecular disulfide bond formation and loss of kinase activity. Pharmacologic inhibition of endogenous reactive oxygen species production in endothelial cells elevated TNIK-dependent ERM phosphorylation, endothelial cell contraction, and cell rounding. Together, we highlight an interplay between TNIK, ERM phosphorylation, and redox signalling in regulating TNF-induced endothelial cell permeability.
Collapse
Affiliation(s)
- Justin Joachim
- School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, BHF Centre of Research Excellence, 125 Coldharbour Lane, King's College London, London SE5 9NU, UK
| | - Davide Maselli
- School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, BHF Centre of Research Excellence, 125 Coldharbour Lane, King's College London, London SE5 9NU, UK
| | - Emmanouela Petsolari
- Randall Centre for Cell and Molecular Biology, King's College London, London SE1 1UL, UK
| | - Jurjan Aman
- Department of Pulmonary Medicine, Amsterdam University Medical Center, location VUMC, Amsterdam, The Netherlands
| | - Pamela Swiatlowska
- Myocardial Function, National Heart and Lung Institute, Imperial College London, ICTEM, Hammersmith Hospital, London, UK
| | - David Killock
- School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, BHF Centre of Research Excellence, 125 Coldharbour Lane, King's College London, London SE5 9NU, UK
| | - Hiba Chaudhry
- School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, BHF Centre of Research Excellence, 125 Coldharbour Lane, King's College London, London SE5 9NU, UK
| | - Ali A Zarban
- School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, BHF Centre of Research Excellence, 125 Coldharbour Lane, King's College London, London SE5 9NU, UK
- Department of Pharmacological Sciences, Faculty of Pharmacy, Jazan University, Saudi Arabia
| | - Mosharraf Sarker
- Pharmacy and Biomolecular Sciences, Faculty of Science, Liverpool John Moores University, Liverpool, UK
| | - Paul Fraser
- School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, BHF Centre of Research Excellence, 125 Coldharbour Lane, King's College London, London SE5 9NU, UK
| | - Simon J Cleary
- Institute of Pharmaceutical Science, King's College London, Floor 5, Southwark Wing, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Richard Amison
- School of Cancer and Pharmaceutical Sciences, Pulmonary Pharmacology Unit, King's College London, London, UK
| | - Isabelle Cuthbert
- School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, BHF Centre of Research Excellence, 125 Coldharbour Lane, King's College London, London SE5 9NU, UK
| | - Yue Yang
- School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, BHF Centre of Research Excellence, 125 Coldharbour Lane, King's College London, London SE5 9NU, UK
| | - Magda Meier
- School of Genetics and Genomic Medicine, University College London Institute of Child Health, London, UK
| | - Franca Fraternali
- Randall Centre for Cell and Molecular Biology, King's College London, London SE1 1UL, UK
- Division of Biosciences, Structural and Molecular Biology Department, University College London, Darwin (SMB) Building, Gower Street, London WC1E 6BT, UK
- Department of Structural and Molecular Biology, Division of Biosciences and Institute of Structural and Molecular Biology, University College London, London WC1E 6BT, UK Department of Biological Sciences, Birkbeck, University of London, London WC1E 7HX, United Kingdom
| | - Susan D Brain
- School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, BHF Centre of Research Excellence, 125 Coldharbour Lane, King's College London, London SE5 9NU, UK
| | - Ajay M Shah
- School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, BHF Centre of Research Excellence, 125 Coldharbour Lane, King's College London, London SE5 9NU, UK
| | - Aleksandar Ivetic
- School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, BHF Centre of Research Excellence, 125 Coldharbour Lane, King's College London, London SE5 9NU, UK
| |
Collapse
|
4
|
Liu X, Li Y, Zhang W, Gao N, Chen J, Xiao C, Zhang G. Inhibition of cIAP1/2 reduces RIPK1 phosphorylation in pulmonary endothelial cells and alleviate sepsis-induced lung injury and inflammatory response. Immunol Res 2024; 72:841-850. [PMID: 38748318 DOI: 10.1007/s12026-024-09491-8] [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: 02/03/2024] [Accepted: 05/08/2024] [Indexed: 06/01/2024]
Abstract
Acute respiratory distress syndrome (ARDS)/acute lung injury (ALI) is a severe complication of sepsis characterized by acute respiratory distress, hypoxemia, and diffuse bilateral pulmonary infiltrates. The regulation of RIPK1 is an important part of the inflammatory response, and cIAP1/2 serves as the E3 ubiquitin ligase for RIPK1. In this study, we investigated the effect and mechanism of cIAP1/2 inhibition on sepsis-induced lung injury. Our results showed that cIAP1/2 inhibition can alleviate sepsis-induced lung injury and reduce the inflammatory response, which is accompanied by downregulation of RIPK1 phosphorylation and ubiquitination. Additionally, cIAP1/2 inhibition led to the up-regulation of programmed cell death, including apoptosis, necroptosis, and pyroptosis, and inhibiting these three cell death pathways can further reduce the inflammatory response, which is similar to the recently discovered programmed cell death pathway PANoptosis. Our findings suggest that cIAP1/2 and PANoptosis inhibition may be a new strategy for treating sepsis-induced lung injury and provide important references for further exploring the mechanism of sepsis-induced lung injury and identifying new therapeutic targets.
Collapse
Affiliation(s)
- Xiaoyu Liu
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Emergency, China-Japan Friendship Hospital, Beijing, China
| | - Yan Li
- Department of Emergency, China-Japan Friendship Hospital, Beijing, China
| | - Weijian Zhang
- Department of Emergency, China-Japan Friendship Hospital, Beijing, China
- Peking University, China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Nan Gao
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Emergency, China-Japan Friendship Hospital, Beijing, China
| | - Jie Chen
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Emergency, China-Japan Friendship Hospital, Beijing, China
| | - Cheng Xiao
- Department of Emergency, China-Japan Friendship Hospital, Beijing, China.
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China.
| | - Guoqiang Zhang
- Department of Emergency, China-Japan Friendship Hospital, Beijing, China.
| |
Collapse
|
5
|
Larionov A, Hammer CM, Fiedler K, Filgueira L. Dynamics of Endothelial Cell Diversity and Plasticity in Health and Disease. Cells 2024; 13:1276. [PMID: 39120307 PMCID: PMC11312403 DOI: 10.3390/cells13151276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/19/2024] [Accepted: 07/19/2024] [Indexed: 08/10/2024] Open
Abstract
Endothelial cells (ECs) are vital structural units of the cardiovascular system possessing two principal distinctive properties: heterogeneity and plasticity. Endothelial heterogeneity is defined by differences in tissue-specific endothelial phenotypes and their high predisposition to modification along the length of the vascular bed. This aspect of heterogeneity is closely associated with plasticity, the ability of ECs to adapt to environmental cues through the mobilization of genetic, molecular, and structural alterations. The specific endothelial cytoarchitectonics facilitate a quick structural cell reorganization and, furthermore, easy adaptation to the extrinsic and intrinsic environmental stimuli, known as the epigenetic landscape. ECs, as universally distributed and ubiquitous cells of the human body, play a role that extends far beyond their structural function in the cardiovascular system. They play a crucial role in terms of barrier function, cell-to-cell communication, and a myriad of physiological and pathologic processes. These include development, ontogenesis, disease initiation, and progression, as well as growth, regeneration, and repair. Despite substantial progress in the understanding of endothelial cell biology, the role of ECs in healthy conditions and pathologies remains a fascinating area of exploration. This review aims to summarize knowledge and concepts in endothelial biology. It focuses on the development and functional characteristics of endothelial cells in health and pathological conditions, with a particular emphasis on endothelial phenotypic and functional heterogeneity.
Collapse
Affiliation(s)
- Alexey Larionov
- Faculty of Science and Medicine, Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700 Fribourg, Switzerland; (C.M.H.); (L.F.)
| | - Christian Manfred Hammer
- Faculty of Science and Medicine, Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700 Fribourg, Switzerland; (C.M.H.); (L.F.)
| | - Klaus Fiedler
- Independent Researcher, CH-1700 Fribourg, Switzerland;
| | - Luis Filgueira
- Faculty of Science and Medicine, Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700 Fribourg, Switzerland; (C.M.H.); (L.F.)
| |
Collapse
|
6
|
Wei L, Dankwa S, Vijayan K, Smith JD, Kaushansky A. Interrogating endothelial barrier regulation by temporally resolved kinase network generation. Life Sci Alliance 2024; 7:e202302522. [PMID: 38467420 PMCID: PMC10927359 DOI: 10.26508/lsa.202302522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 03/13/2024] Open
Abstract
Kinases are key players in endothelial barrier regulation, yet their temporal function and regulatory phosphosignaling networks are incompletely understood. We developed a novel methodology, Temporally REsolved KInase Network Generation (TREKING), which combines a 28-kinase inhibitor screen with machine learning and network reconstruction to build time-resolved, functional phosphosignaling networks. We demonstrated the utility of TREKING for identifying pathways mediating barrier integrity after activation by thrombin with or without TNF preconditioning in brain endothelial cells. TREKING predicted over 100 kinases involved in barrier regulation and discerned complex condition-specific pathways. For instance, the MAPK-activated protein kinase 2 (MAPKAPK2/MK2) had early barrier-weakening activity in both inflammatory conditions but late barrier-strengthening activity exclusively with thrombin alone. Using temporal Western blotting, we confirmed that MAPKAPK2/MK2 was differentially phosphorylated under the two inflammatory conditions. We further showed with lentivirus-mediated knockdown of MAPK14/p38α and drug targeting the MAPK14/p38α-MAPKAPK2/MK2 complex that a MAP3K20/ZAK-MAPK14/p38α axis controlled the late activation of MAPKAPK2/MK2 in the thrombin-alone condition. Beyond the MAPKAPK2/MK2 switch, TREKING predicts extensive interconnected networks that control endothelial barrier dynamics.
Collapse
Affiliation(s)
- Ling Wei
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Selasi Dankwa
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Kamalakannan Vijayan
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Joseph D Smith
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Alexis Kaushansky
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
- Department of Global Health, University of Washington, Seattle, WA, USA
| |
Collapse
|
7
|
Hooglugt A, van der Stoel MM, Shapeti A, Neep BF, de Haan A, van Oosterwyck H, Boon RA, Huveneers S. DLC1 promotes mechanotransductive feedback for YAP via RhoGAP-mediated focal adhesion turnover. J Cell Sci 2024; 137:jcs261687. [PMID: 38563084 PMCID: PMC11112125 DOI: 10.1242/jcs.261687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 03/25/2024] [Indexed: 04/04/2024] Open
Abstract
Angiogenesis is a tightly controlled dynamic process demanding a delicate equilibrium between pro-angiogenic signals and factors that promote vascular stability. The spatiotemporal activation of the transcriptional co-factors YAP (herein referring to YAP1) and TAZ (also known WWTR1), collectively denoted YAP/TAZ, is crucial to allow for efficient collective endothelial migration in angiogenesis. The focal adhesion protein deleted-in-liver-cancer-1 (DLC1) was recently described as a transcriptional downstream target of YAP/TAZ in endothelial cells. In this study, we uncover a negative feedback loop between DLC1 expression and YAP activity during collective migration and sprouting angiogenesis. In particular, our study demonstrates that signaling via the RhoGAP domain of DLC1 reduces nuclear localization of YAP and its transcriptional activity. Moreover, the RhoGAP activity of DLC1 is essential for YAP-mediated cellular processes, including the regulation of focal adhesion turnover, traction forces, and sprouting angiogenesis. We show that DLC1 restricts intracellular cytoskeletal tension by inhibiting Rho signaling at the basal adhesion plane, consequently reducing nuclear YAP localization. Collectively, these findings underscore the significance of DLC1 expression levels and its function in mitigating intracellular tension as a pivotal mechanotransductive feedback mechanism that finely tunes YAP activity throughout the process of sprouting angiogenesis.
Collapse
Affiliation(s)
- Aukie Hooglugt
- Amsterdam UMC, University of Amsterdam, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, 1105AZ Amsterdam, the Netherlands
- Amsterdam UMC, VU University Medical Center, Department of Physiology, Amsterdam Cardiovascular Sciences, 1081HZ Amsterdam, the Netherlands
| | - Miesje M. van der Stoel
- Amsterdam UMC, University of Amsterdam, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, 1105AZ Amsterdam, the Netherlands
| | - Apeksha Shapeti
- KU Leuven, Department of Mechanical Engineering, Biomechanics section, 3001 Leuven, Belgium
| | - Beau F. Neep
- Amsterdam UMC, University of Amsterdam, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, 1105AZ Amsterdam, the Netherlands
- Amsterdam UMC, VU University Medical Center, Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, 1081HZ Amsterdam, the Netherlands
| | - Annett de Haan
- Amsterdam UMC, University of Amsterdam, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, 1105AZ Amsterdam, the Netherlands
| | - Hans van Oosterwyck
- KU Leuven, Department of Mechanical Engineering, Biomechanics section, 3001 Leuven, Belgium
- KU Leuven, Prometheus, Division of Skeletal Tissue Engineering, 3000 Leuven, Belgium
| | - Reinier A. Boon
- Amsterdam UMC, VU University Medical Center, Department of Physiology, Amsterdam Cardiovascular Sciences, 1081HZ Amsterdam, the Netherlands
- German Center for Cardiovascular Research (DZHK), Partner Site Rhein-Main, 60590 Frankfurt am Main, Germany
- Goethe University, Institute of Cardiovascular Regeneration, 60590 Frankfurt am Main, Germany
| | - Stephan Huveneers
- Amsterdam UMC, University of Amsterdam, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, 1105AZ Amsterdam, the Netherlands
| |
Collapse
|
8
|
Juin A, Spence HJ, Machesky LM. Dichotomous role of the serine/threonine kinase MAP4K4 in pancreatic ductal adenocarcinoma onset and metastasis through control of AKT and ERK pathways. J Pathol 2024; 262:454-466. [PMID: 38229581 DOI: 10.1002/path.6248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 10/24/2023] [Accepted: 12/04/2023] [Indexed: 01/18/2024]
Abstract
MAP4K4 is a serine/threonine kinase of the STE20 family involved in the regulation of actin cytoskeleton dynamics and cell motility. It has been proposed as a target of angiogenesis and inhibitors show potential in cardioprotection. MAP4K4 also mediates cell invasion in vitro, is overexpressed in various types of cancer, and is associated with poor patient prognosis. Recently, MAP4K4 has been shown to be overexpressed in pancreatic cancer, but its role in tumour initiation, progression, and metastasis is unknown. Here, using the KrasG12D Trp53R172H Pdx1-Cre (KPC) mouse model of pancreatic ductal adenocarcinoma (PDAC), we show that deletion of Map4k4 drives tumour initiation and progression. Moreover, we report that the acceleration of tumour onset is also associated with an overactivation of ERK and AKT, two major downstream effectors of KRAS, in vitro and in vivo. In contrast to the accelerated tumour onset caused by loss of MAP4K4, we observed a reduction in metastatic burden with both the KPC model and in an intraperitoneal transplant assay indicating a major role of MAP4K4 in metastatic seeding. In summary, our study sheds light on the dichotomous role of MAP4K4 in the initiation of PDAC onset, progression, and metastatic dissemination. It also identifies MAP4K4 as a possible druggable target against pancreatic cancer spread, but with the caveat that targeting MAP4K4 might accelerate early tumorigenesis. © 2024 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
| | | | - Laura M Machesky
- CRUK Beatson Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| |
Collapse
|
9
|
Duijvelaar E, Gisby J, Peters JE, Bogaard HJ, Aman J. Longitudinal plasma proteomics reveals biomarkers of alveolar-capillary barrier disruption in critically ill COVID-19 patients. Nat Commun 2024; 15:744. [PMID: 38272877 PMCID: PMC10811341 DOI: 10.1038/s41467-024-44986-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 01/11/2024] [Indexed: 01/27/2024] Open
Abstract
The pathobiology of respiratory failure in COVID-19 consists of a complex interplay between viral cytopathic effects and a dysregulated host immune response. In critically ill patients, imatinib treatment demonstrated potential for reducing invasive ventilation duration and mortality. Here, we perform longitudinal profiling of 6385 plasma proteins in 318 hospitalised patients to investigate the biological processes involved in critical COVID-19, and assess the effects of imatinib treatment. Nine proteins measured at hospital admission accurately predict critical illness development. Next to dysregulation of inflammation, critical illness is characterised by pathways involving cellular adhesion, extracellular matrix turnover and tissue remodelling. Imatinib treatment attenuates protein perturbations associated with inflammation and extracellular matrix turnover. These proteomic alterations are contextualised using external pulmonary RNA-sequencing data of deceased COVID-19 patients and imatinib-treated Syrian hamsters. Together, we show that alveolar capillary barrier disruption in critical COVID-19 is reflected in the plasma proteome, and is attenuated with imatinib treatment. This study comprises a secondary analysis of both clinical data and plasma samples derived from a clinical trial that was registered with the EU Clinical Trials Register (EudraCT 2020-001236-10, https://www.clinicaltrialsregister.eu/ctr-search/trial/2020-001236-10/NL ) and Netherlands Trial Register (NL8491, https://www.trialregister.nl/trial/8491 ).
Collapse
Affiliation(s)
- Erik Duijvelaar
- Department of Pulmonary Medicine, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands.
| | - Jack Gisby
- Department of Immunology and Inflammation, Centre for Inflammatory Disease, Imperial College London, London, UK
| | - James E Peters
- Department of Immunology and Inflammation, Centre for Inflammatory Disease, Imperial College London, London, UK
| | - Harm Jan Bogaard
- Department of Pulmonary Medicine, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Jurjan Aman
- Department of Pulmonary Medicine, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands.
| |
Collapse
|
10
|
Geyer CE, Chen HJ, Bye AP, Manz XD, Guerra D, Caniels TG, Bijl TP, Griffith GR, Hoepel W, de Taeye SW, Veth J, Vlaar AP, Vidarsson G, Bogaard HJ, Aman J, Gibbins JM, van Gils MJ, de Winther MP, den Dunnen J. Identification of new drugs to counteract anti-spike IgG-induced hyperinflammation in severe COVID-19. Life Sci Alliance 2023; 6:e202302106. [PMID: 37699657 PMCID: PMC10497933 DOI: 10.26508/lsa.202302106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 08/24/2023] [Accepted: 08/29/2023] [Indexed: 09/14/2023] Open
Abstract
Previously, we and others have shown that SARS-CoV-2 spike-specific IgG antibodies play a major role in disease severity in COVID-19 by triggering macrophage hyperactivation, disrupting endothelial barrier integrity, and inducing thrombus formation. This hyperinflammation is dependent on high levels of anti-spike IgG with aberrant Fc tail glycosylation, leading to Fcγ receptor hyperactivation. For development of immune-regulatory therapeutics, drug specificity is crucial to counteract excessive inflammation whereas simultaneously minimizing the inhibition of antiviral immunity. We here developed an in vitro activation assay to screen for small molecule drugs that specifically counteract antibody-induced pathology. We identified that anti-spike-induced inflammation is specifically blocked by small molecule inhibitors against SYK and PI3K. We identified SYK inhibitor entospletinib as the most promising candidate drug, which also counteracted anti-spike-induced endothelial dysfunction and thrombus formation. Moreover, entospletinib blocked inflammation by different SARS-CoV-2 variants of concern. Combined, these data identify entospletinib as a promising treatment for severe COVID-19.
Collapse
Affiliation(s)
- Chiara E Geyer
- Center for Experimental and Molecular Medicine, Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Hung-Jen Chen
- Center for Experimental and Molecular Medicine, Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Alexander P Bye
- Institute for Cardiovascular and Metabolic Research, and School of Biological Sciences, University of Reading, Reading, UK
- Molecular and Clinical Sciences Research Institute, St George's University, London, UK
- School of Pharmacy, University of Reading, Reading, UK
| | - Xue D Manz
- Pulmonary Medicine, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Denise Guerra
- Medical Microbiology and Infection Prevention, Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Tom G Caniels
- Medical Microbiology and Infection Prevention, Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Tom Pl Bijl
- Medical Microbiology and Infection Prevention, Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Guillermo R Griffith
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Willianne Hoepel
- Center for Experimental and Molecular Medicine, Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Steven W de Taeye
- Medical Microbiology and Infection Prevention, Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Jennifer Veth
- Center for Experimental and Molecular Medicine, Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Alexander Pj Vlaar
- Department of Intensive Care Medicine, Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Gestur Vidarsson
- Experimental Immunohematology, Sanquin Research, Amsterdam, Netherlands
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
| | - Harm Jan Bogaard
- Pulmonary Medicine, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Jurjan Aman
- Pulmonary Medicine, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Jonathan M Gibbins
- Institute for Cardiovascular and Metabolic Research, and School of Biological Sciences, University of Reading, Reading, UK
| | - Marit J van Gils
- Medical Microbiology and Infection Prevention, Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Menno Pj de Winther
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Jeroen den Dunnen
- Center for Experimental and Molecular Medicine, Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Centers, Amsterdam, Netherlands
| |
Collapse
|
11
|
Lv L, Cui EH, Wang B, Li LQ, Hua F, Lu HD, Chen N, Chen WY. Multiomics reveal human umbilical cord mesenchymal stem cells improving acute lung injury via the lung-gut axis. World J Stem Cells 2023; 15:908-930. [PMID: 37900940 PMCID: PMC10600741 DOI: 10.4252/wjsc.v15.i9.908] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/23/2023] [Accepted: 09/06/2023] [Indexed: 09/25/2023] Open
Abstract
BACKGROUND Acute lung injury (ALI) and its final severe stage, acute respiratory distress syndrome, are associated with high morbidity and mortality rates in patients due to the lack of effective specific treatments. Gut microbiota homeostasis, including that in ALI, is important for human health. Evidence suggests that the gut microbiota improves lung injury through the lung-gut axis. Human umbilical cord mesenchymal cells (HUC-MSCs) have attractive prospects for ALI treatment. This study hypothesized that HUC-MSCs improve ALI via the lung-gut microflora. AIM To explore the effects of HUC-MSCs on lipopolysaccharide (LPS)-induced ALI in mice and the involvement of the lung-gut axis in this process. METHODS C57BL/6 mice were randomly divided into four groups (18 rats per group): Sham, sham + HUC-MSCs, LPS, and LPS + HUC-MSCs. ALI was induced in mice by intraperitoneal injections of LPS (10 mg/kg). After 6 h, mice were intervened with 0.5 mL phosphate buffered saline (PBS) containing 1 × 106 HUC-MSCs by intraperitoneal injections. For the negative control, 100 mL 0.9% NaCl and 0.5 mL PBS were used. Bronchoalveolar lavage fluid (BALF) was obtained from anesthetized mice, and their blood, lungs, ileum, and feces were obtained by an aseptic technique following CO2 euthanasia. Wright's staining, enzyme-linked immunosorbent assay, hematoxylin-eosin staining, Evans blue dye leakage assay, immunohistochemistry, fluorescence in situ hybridization, western blot, 16S rDNA sequencing, and non-targeted metabolomics were used to observe the effect of HUC-MSCs on ALI mice, and the involvement of the lung-gut axis in this process was explored. One-way analysis of variance with post-hoc Tukey's test, independent-sample Student's t-test, Wilcoxon rank-sum test, and Pearson correlation analysis were used for statistical analyses. RESULTS HUC-MSCs were observed to improve pulmonary edema and lung and ileal injury, and decrease mononuclear cell and neutrophil counts, protein concentrations in BALF and inflammatory cytokine levels in the serum, lung, and ileum of ALI mice. Especially, HUC-MSCs decreased Evans blue concentration and Toll-like receptor 4, myeloid differentiation factor 88, p-nuclear factor kappa-B (NF-κB)/NF-κB, and p-inhibitor α of NF-κB (p-IκBα)/IκBα expression levels in the lung, and raised the pulmonary vascular endothelial-cadherin, zonula occludens-1 (ZO-1), and occludin levels and ileal ZO-1, claudin-1, and occludin expression levels. HUC-MSCs improved gut and BALF microbial homeostases. The number of pathogenic bacteria decreased in the BALF of ALI mice treated with HUC-MSCs. Concurrently, the abundances of Oscillospira and Coprococcus in the feces of HUS-MSC-treated ALI mice were significantly increased. In addition, Lactobacillus, Bacteroides, and unidentified_Rikenellaceae genera appeared in both feces and BALF. Moreover, this study performed metabolomic analysis on the lung tissue and identified five upregulated metabolites and 11 downregulated metabolites in the LPS + MSC group compared to the LPS group, which were related to the purine metabolism and the taste transduction signaling pathways. Therefore, an intrinsic link between lung metabolite levels and BALF flora homeostasis was established. CONCLUSION This study suggests that HUM-MSCs attenuate ALI by redefining the gut and lung microbiota.
Collapse
Affiliation(s)
- Lu Lv
- Department of Respiratory and Critical Care Medicine, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou 313000, Zhejiang Province, China
| | - En-Hai Cui
- Department of Respiratory and Critical Care Medicine, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou 313000, Zhejiang Province, China.
| | - Bin Wang
- Department of Respiratory and Critical Care Medicine, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou 313000, Zhejiang Province, China
| | - Li-Qin Li
- Traditional Chinese Medicine Key Laboratory Cultivation Base of Zhejiang Province for the Development and Clinical Transformation of Immunomodulatory Drugs, Huzhou 313000, Zhejiang Province, China
| | - Feng Hua
- Department of Respiratory and Critical Care Medicine, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou 313000, Zhejiang Province, China
| | - Hua-Dong Lu
- Department of Respiratory and Critical Care Medicine, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou 313000, Zhejiang Province, China
| | - Na Chen
- Department of Respiratory and Critical Care Medicine, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou 313000, Zhejiang Province, China
| | - Wen-Yan Chen
- Department of Respiratory and Critical Care Medicine, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou 313000, Zhejiang Province, China
| |
Collapse
|
12
|
Alberici Delsin LE, Plutoni C, Clouvel A, Keil S, Marpeaux L, Elouassouli L, Khavari A, Ehrlicher AJ, Emery G. MAP4K4 regulates forces at cell-cell and cell-matrix adhesions to promote collective cell migration. Life Sci Alliance 2023; 6:e202302196. [PMID: 37369604 PMCID: PMC10300198 DOI: 10.26508/lsa.202302196] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/12/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
Collective cell migration is not only important for development and tissue homeostasis but can also promote cancer metastasis. To migrate collectively, cells need to coordinate cellular extensions and retractions, adhesion sites dynamics, and forces generation and transmission. Nevertheless, the regulatory mechanisms coordinating these processes remain elusive. Using A431 carcinoma cells, we identify the kinase MAP4K4 as a central regulator of collective migration. We show that MAP4K4 inactivation blocks the migration of clusters, whereas its overexpression decreases cluster cohesion. MAP4K4 regulates protrusion and retraction dynamics, remodels the actomyosin cytoskeleton, and controls the stability of both cell-cell and cell-substrate adhesion. MAP4K4 promotes focal adhesion disassembly through the phosphorylation of the actin and plasma membrane crosslinker moesin but disassembles adherens junctions through a moesin-independent mechanism. By analyzing traction and intercellular forces, we found that MAP4K4 loss of function leads to a tensional disequilibrium throughout the cell cluster, increasing the traction forces and the tension loading at the cell-cell adhesions. Together, our results indicate that MAP4K4 activity is a key regulator of biomechanical forces at adhesion sites, promoting collective migration.
Collapse
Affiliation(s)
- Lara Elis Alberici Delsin
- Vesicular Trafficking and Cell Signalling Research Unit, Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, Canada
| | - Cédric Plutoni
- Vesicular Trafficking and Cell Signalling Research Unit, Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, Canada
| | - Anna Clouvel
- Department of Bioengineering, McGill University, Montreal, Canada
| | - Sarah Keil
- Vesicular Trafficking and Cell Signalling Research Unit, Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, Canada
| | - Léa Marpeaux
- Vesicular Trafficking and Cell Signalling Research Unit, Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, Canada
| | - Lina Elouassouli
- Vesicular Trafficking and Cell Signalling Research Unit, Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, Canada
| | - Adele Khavari
- Department of Bioengineering, McGill University, Montreal, Canada
| | | | - Gregory Emery
- Vesicular Trafficking and Cell Signalling Research Unit, Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, Canada
- Department of Pathology and Cell Biology, Faculty of Medicine, Université de Montréal, Montréal, Canada
| |
Collapse
|
13
|
Abstract
The endothelium is a dynamic, semipermeable layer lining all blood vessels, regulating blood vessel formation and barrier function. Proper composition and function of the endothelial barrier are required for fluid homeostasis, and clinical conditions characterized by barrier disruption are associated with severe morbidity and high mortality rates. Endothelial barrier properties are regulated by cell-cell junctions and intracellular signaling pathways governing the cytoskeleton, but recent insights indicate an increasingly important role for integrin-mediated cell-matrix adhesion and signaling in endothelial barrier regulation. Here, we discuss diseases characterized by endothelial barrier disruption, and provide an overview of the composition of endothelial cell-matrix adhesion complexes and associated signaling pathways, their crosstalk with cell-cell junctions, and with other receptors. We further present recent insights into the role of cell-matrix adhesions in the developing and mature/adult endothelium of various vascular beds, and discuss how the dynamic regulation and turnover of cell-matrix adhesions regulates endothelial barrier function in (patho)physiological conditions like angiogenesis, inflammation and in response to hemodynamic stress. Finally, as clinical conditions associated with vascular leak still lack direct treatment, we focus on how understanding of endothelial cell-matrix adhesion may provide novel targets for treatment, and discuss current translational challenges and future perspectives.
Collapse
Affiliation(s)
- Jurjan Aman
- Department of Pulmonology, Amsterdam University Medical Center, the Netherlands (J.A.)
| | - Coert Margadant
- Department of Medical Oncology, Amsterdam University Medical Center, the NetherlandsInstitute of Biology, Leiden University, the Netherlands (C.M.)
| |
Collapse
|
14
|
Jovanovic D, Yan S, Baumgartner M. The molecular basis of the dichotomous functionality of MAP4K4 in proliferation and cell motility control in cancer. Front Oncol 2022; 12:1059513. [PMID: 36568222 PMCID: PMC9774001 DOI: 10.3389/fonc.2022.1059513] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 11/15/2022] [Indexed: 12/13/2022] Open
Abstract
The finely tuned integration of intra- and extracellular cues by components of the mitogen-activated protein kinase (MAPK) signaling pathways controls the mutually exclusive phenotypic manifestations of uncontrolled growth and tumor cell dissemination. The Ser/Thr kinase MAP4K4 is an upstream integrator of extracellular cues involved in both proliferation and cell motility control. Initially identified as an activator of the c-Jun N-terminal kinase (JNK), the discovery of diverse functions and additional effectors of MAP4K4 beyond JNK signaling has considerably broadened our understanding of this complex kinase. The implication of MAP4K4 in the regulation of cytoskeleton dynamics and cell motility provided essential insights into its role as a pro-metastatic kinase in cancer. However, the more recently revealed role of MAP4K4 as an activator of the Hippo tumor suppressor pathway has complicated the understanding of MAP4K4 as an oncogenic driver kinase. To develop a better understanding of the diverse functions of MAP4K4 and their potential significance in oncogenesis and tumor progression, we have collected and assessed the current evidence of MAP4K4 implication in molecular mechanisms that control proliferation and promote cell motility. A better understanding of these mechanisms is particularly relevant in the brain, where MAP4K4 is highly expressed and under pathological conditions either drives neuronal cell death in neurodegenerative diseases or cell dissemination in malignant tumors. We review established effectors and present novel interactors of MAP4K4, which offer mechanistic insights into MAP4K4 function and may inspire novel intervention strategies. We discuss possible implications of novel interactors in tumor growth and dissemination and evaluate potential therapeutic strategies to selectively repress pro-oncogenic functions of MAP4K4.
Collapse
Affiliation(s)
| | | | - Martin Baumgartner
- Pediatric Molecular Neuro-Oncology Research, Children’s Research Centre, Division of Oncology, University Children’s Hospital Zürich, Zürich, Switzerland
| |
Collapse
|
15
|
Milenkovic D, Rodriguez‐Mateos A, Lucosz M, Istas G, Declerck K, Sansone R, Deenen R, Köhrer K, Corral‐Jara KF, Altschmied J, Haendeler J, Kelm M, Berghe WV, Heiss C. Flavanol Consumption in Healthy Men Preserves Integrity of Immunological-Endothelial Barrier Cell Functions: Nutri(epi)genomic Analysis. Mol Nutr Food Res 2022; 66:e2100991. [PMID: 35094491 PMCID: PMC9787825 DOI: 10.1002/mnfr.202100991] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 01/16/2022] [Indexed: 12/30/2022]
Abstract
SCOPE While cocoa flavanol (CF) consumption improves cardiovascular risk biomarkers, molecular mechanisms underlying their protective effects are not understood. OBJECTIVE To investigate nutri(epi)genomic effects of CF and identify regulatory networks potential mediating vascular health benefits. METHODS AND RESULTS Twenty healthy middle-aged men consume CF (bi-daily 450 mg) or control drinks for 1 month. Microarray analysis identifies 2235 differentially expressed genes (DEG) involved in processes regulating immune response, cell adhesion, or cytoskeleton organization. Distinct patterns of DEG correlate with CF-related changes in endothelial function, arterial stiffness, and blood pressure. DEG profile negatively correlates with expression profiles of cardiovascular disease patients. CF modulated DNA methylation profile of genes implicates in cell adhesion, actin cytoskeleton organization, or cell signaling. In silico docking analyses indicate that CF metabolites have the potential of binding to cell signaling proteins and transcription factors. Incubation of plasma obtained after CF consumption decrease monocyte to endothelial adhesion and dose-dependently increase nitric oxide-dependent chemotaxis of circulating angiogenic cells further validating the biological functions of CF metabolites. CONCLUSION In healthy humans, CF consumption may mediate vascular protective effects by modulating gene expression and DNA methylation towards a cardiovascular protective effect, in agreement with clinical results, by preserving integrity of immunological-endothelial barrier functions.
Collapse
Affiliation(s)
- Dragan Milenkovic
- Department of NutritionUniversity of California DavisDavisCA95616USA
- INRAEUNHUniversité Clermont AuvergneClermont‐FerrandF‐63000France
| | - Ana Rodriguez‐Mateos
- Division of CardiologyPulmonology, and Vascular MedicineMedical FacultyUniversity Hospital DüsseldorfDüsseldorfGermany
- Department of Nutritional SciencesSchool of Life Course and Population SciencesFaculty of Life Sciences and MedicineKing's College LondonLondonUK
| | - Margarete Lucosz
- Division of CardiologyPulmonology, and Vascular MedicineMedical FacultyUniversity Hospital DüsseldorfDüsseldorfGermany
| | - Geoffrey Istas
- Division of CardiologyPulmonology, and Vascular MedicineMedical FacultyUniversity Hospital DüsseldorfDüsseldorfGermany
- Department of Nutritional SciencesSchool of Life Course and Population SciencesFaculty of Life Sciences and MedicineKing's College LondonLondonUK
| | - Ken Declerck
- PPESDepartment of Biomedical SciencesUniversity of Antwerp (UA)WilrijkBelgium
| | - Roberto Sansone
- Division of CardiologyPulmonology, and Vascular MedicineMedical FacultyUniversity Hospital DüsseldorfDüsseldorfGermany
| | - René Deenen
- Biological and Medical Research Center (BMFZ)Heinrich Heine UniversityDüsseldorfGermany
| | - Karl Köhrer
- Biological and Medical Research Center (BMFZ)Heinrich Heine UniversityDüsseldorfGermany
| | | | - Joachim Altschmied
- Environmentally‐induced Cardiovascular DegenerationClinical Chemistry and Laboratory DiagnosticsMedical FacultyUniversity Hospital and Heinrich‐Heine UniversityDüsseldorfGermany
- IUF‐Leibniz Research Institute for Environmental MedicineDüsseldorfGermany
| | - Judith Haendeler
- Environmentally‐induced Cardiovascular DegenerationClinical Chemistry and Laboratory DiagnosticsMedical FacultyUniversity Hospital and Heinrich‐Heine UniversityDüsseldorfGermany
| | - Malte Kelm
- Division of CardiologyPulmonology, and Vascular MedicineMedical FacultyUniversity Hospital DüsseldorfDüsseldorfGermany
| | - Wim Vanden Berghe
- PPESDepartment of Biomedical SciencesUniversity of Antwerp (UA)WilrijkBelgium
| | - Christian Heiss
- Division of CardiologyPulmonology, and Vascular MedicineMedical FacultyUniversity Hospital DüsseldorfDüsseldorfGermany
- Clinical Medicine SectionDepartment of Clinical and Experimental MedicineFaculty of Health and Medical SciencesUniversity of SurreyGuildfordUK
- Department of Vascular MedicineSurrey and Sussex NHS Healthcare TrustEast Surrey HospitalRedhillUK
| |
Collapse
|
16
|
Dankwa S, Dols MM, Wei L, Glennon EKK, Kain HS, Kaushansky A, Smith JD. Exploiting polypharmacology to dissect host kinases and kinase inhibitors that modulate endothelial barrier integrity. Cell Chem Biol 2021; 28:1679-1692.e4. [PMID: 34216546 PMCID: PMC8688180 DOI: 10.1016/j.chembiol.2021.06.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 04/29/2021] [Accepted: 06/09/2021] [Indexed: 10/21/2022]
Abstract
Kinase inhibitors are promising drugs to stabilize the endothelial barrier following inflammatory damage. However, our limited knowledge of how kinase signaling activates barrier-restorative pathways and the complexity of multi-target drugs have hindered drug discovery and repurposing efforts. Here, we apply a kinase regression approach that exploits drug polypharmacology to investigate endothelial barrier regulation. A screen of 28 kinase inhibitors identified multiple inhibitors that promote endothelial barrier integrity and revealed divergent barrier phenotypes for BCR-ABL drugs. Target deconvolution predicted 50 barrier-regulating kinases from diverse kinase families. Using gene knockdowns, we identified kinases with a role in endothelial barrier regulation and dissected different mechanisms of action of barrier-protective kinase inhibitors. These results demonstrate the importance of polypharmacology in the endothelial barrier phenotype of kinase inhibitors and provide promising new leads for barrier-strengthening therapies.
Collapse
Affiliation(s)
- Selasi Dankwa
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Mary-Margaret Dols
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Ling Wei
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Elizabeth K K Glennon
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Heather S Kain
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Alexis Kaushansky
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA; Department of Pediatrics, University of Washington, Seattle, WA 98105, USA.
| | - Joseph D Smith
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA; Department of Pediatrics, University of Washington, Seattle, WA 98105, USA.
| |
Collapse
|
17
|
Hernandez SJ, Fote G, Reyes-Ortiz AM, Steffan JS, Thompson LM. Cooperation of cell adhesion and autophagy in the brain: Functional roles in development and neurodegenerative disease. Matrix Biol Plus 2021; 12:100089. [PMID: 34786551 PMCID: PMC8579148 DOI: 10.1016/j.mbplus.2021.100089] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/11/2021] [Accepted: 10/18/2021] [Indexed: 12/19/2022] Open
Abstract
Cellular adhesive connections directed by the extracellular matrix (ECM) and maintenance of cellular homeostasis by autophagy are seemingly disparate functions that are molecularly intertwined, each regulating the other. This is an emerging field in the brain where the interplay between adhesion and autophagy functions at the intersection of neuroprotection and neurodegeneration. The ECM and adhesion proteins regulate autophagic responses to direct protein clearance and guide regenerative programs that go awry in brain disorders. Concomitantly, autophagic flux acts to regulate adhesion dynamics to mediate neurite outgrowth and synaptic plasticity with functional disruption contributed by neurodegenerative disease. This review highlights the cooperative exchange between cellular adhesion and autophagy in the brain during health and disease. As the mechanistic alliance between adhesion and autophagy has been leveraged therapeutically for metastatic disease, understanding overlapping molecular functions that direct the interplay between adhesion and autophagy might uncover therapeutic strategies to correct or compensate for neurodegeneration.
Collapse
Affiliation(s)
- Sarah J. Hernandez
- Neurobiology and Behavior, University of California Irvine, Irvine, CA 92697, USA
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA 92697, USA
| | - Gianna Fote
- Department of Biological Chemistry, University of California Irvine, Irvine, CA 92697, USA
| | - Andrea M. Reyes-Ortiz
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA 92697, USA
- Department of Biological Chemistry, University of California Irvine, Irvine, CA 92697, USA
| | - Joan S. Steffan
- Psychaitry and Human Behavior, University of California Irvine, Irvine, CA 92697, USA
- Institute of Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA 92617, USA
| | - Leslie M. Thompson
- Neurobiology and Behavior, University of California Irvine, Irvine, CA 92697, USA
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA 92697, USA
- Department of Biological Chemistry, University of California Irvine, Irvine, CA 92697, USA
- Psychaitry and Human Behavior, University of California Irvine, Irvine, CA 92697, USA
- Institute of Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA 92617, USA
| |
Collapse
|
18
|
Sirt3 Maintains Microvascular Endothelial Adherens Junction Integrity to Alleviate Sepsis-Induced Lung Inflammation by Modulating the Interaction of VE-Cadherin and β-Catenin. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8978795. [PMID: 34630854 PMCID: PMC8500765 DOI: 10.1155/2021/8978795] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/07/2021] [Accepted: 09/15/2021] [Indexed: 11/21/2022]
Abstract
Inflammatory injury is a hallmark of sepsis-induced acute respiratory distress syndrome (ARDS)/acute lung injury (ALI). However, the mechanisms underlying inflammatory injury remain obscure. Here, we developed the novel strategy to suppress lung inflammation through maintaining microvascular endothelial barrier integrity. VE-cadherin is the main adherens junction protein that interacts with β-catenin and forms a complex. We found that lung inflammation was accompanied by decreased VE-cadherin expression and increased β-catenin activity in animal models and human pulmonary microvascular endothelial cells (HPMECs), illuminating the relationship among VE-cadherin/β-catenin complex, microvascular endothelial barrier integrity, and inflammation. Furthermore, we showed that the VE-cadherin/β-catenin complex dissociated upon lung inflammation, while Sirt3 promoted the stability of such a complex. Sirt3 was decreased during lung inflammation in vivo and in vitro. Sirt3 deficiency not only led to the downregulation of VE-cadherin but also enhanced the transcriptional activity of β-catenin that further increased β-catenin target gene MMP-7 expression, thereby promoting inflammatory factor COX-2 expression. Sirt3 overexpression promoted VE-cadherin expression, inhibited β-catenin transcriptional activity, strengthened the stability of the VE-cadherin/β-catenin complex, and suppressed inflammation in HPMECs. Notably, Sirt3 deficiency significantly damaged microvascular endothelial barrier integrity and intensified lung inflammation in animal model. These results demonstrated the role of Sirt3 in modulating microvascular endothelial barrier integrity to inhibit inflammation. Therefore, strategies that aim at enhancing the stability of endothelial VE-cadherin/β-catenin complex are potentially beneficial for preventing sepsis-induced lung inflammation.
Collapse
|
19
|
Hoepel W, Chen HJ, Geyer CE, Allahverdiyeva S, Manz XD, de Taeye SW, Aman J, Mes L, Steenhuis M, Griffith GR, Bonta PI, Brouwer PJM, Caniels TG, van der Straten K, Golebski K, Jonkers RE, Larsen MD, Linty F, Nouta J, van Roomen CPAA, van Baarle FEHP, van Drunen CM, Wolbink G, Vlaar APJ, de Bree GJ, Sanders RW, Willemsen L, Neele AE, van de Beek D, Rispens T, Wuhrer M, Bogaard HJ, van Gils MJ, Vidarsson G, de Winther M, den Dunnen J. High titers and low fucosylation of early human anti-SARS-CoV-2 IgG promote inflammation by alveolar macrophages. Sci Transl Med 2021; 13:eabf8654. [PMID: 33979301 PMCID: PMC8158960 DOI: 10.1126/scitranslmed.abf8654] [Citation(s) in RCA: 167] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 03/05/2021] [Accepted: 05/04/2021] [Indexed: 12/17/2022]
Abstract
Patients diagnosed with coronavirus disease 2019 (COVID-19) become critically ill primarily around the time of activation of the adaptive immune response. Here, we provide evidence that antibodies play a role in the worsening of disease at the time of seroconversion. We show that early-phase severe acute respiratory distress syndrome coronavirus 2 (SARS-CoV-2) spike protein-specific immunoglobulin G (IgG) in serum of critically ill COVID-19 patients induces excessive inflammatory responses by human alveolar macrophages. We identified that this excessive inflammatory response is dependent on two antibody features that are specific for patients with severe COVID-19. First, inflammation is driven by high titers of anti-spike IgG, a hallmark of severe disease. Second, we found that anti-spike IgG from patients with severe COVID-19 is intrinsically more proinflammatory because of different glycosylation, particularly low fucosylation, of the antibody Fc tail. Low fucosylation of anti-spike IgG was normalized in a few weeks after initial infection with SARS-CoV-2, indicating that the increased antibody-dependent inflammation mainly occurs at the time of seroconversion. We identified Fcγ receptor (FcγR) IIa and FcγRIII as the two primary IgG receptors that are responsible for the induction of key COVID-19-associated cytokines such as interleukin-6 and tumor necrosis factor. In addition, we show that anti-spike IgG-activated human macrophages can subsequently break pulmonary endothelial barrier integrity and induce microvascular thrombosis in vitro. Last, we demonstrate that the inflammatory response induced by anti-spike IgG can be specifically counteracted by fostamatinib, an FDA- and EMA-approved therapeutic small-molecule inhibitor of Syk kinase.
Collapse
Affiliation(s)
- Willianne Hoepel
- Department of Rheumatology and Clinical Immunology, Amsterdam UMC, Amsterdam Rheumatology and Immunology Center, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam Infection and Immunity Institute, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands
| | - Hung-Jen Chen
- Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam Cardiovascular Sciences, Amsterdam Infection and Immunity, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands
| | - Chiara E Geyer
- Department of Rheumatology and Clinical Immunology, Amsterdam UMC, Amsterdam Rheumatology and Immunology Center, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam Infection and Immunity Institute, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands
| | - Sona Allahverdiyeva
- Department of Rheumatology and Clinical Immunology, Amsterdam UMC, Amsterdam Rheumatology and Immunology Center, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam Infection and Immunity Institute, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Infection and Immunity Institute, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands
| | - Xue D Manz
- Department of Pulmonary Medicine, Amsterdam UMC, location VUMC, De Boelelaan 1117, 1081 HV Amsterdam, Netherlands
| | - Steven W de Taeye
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Infection and Immunity Institute, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory Academic Medical Centre, Plesmanlaan 125, 1066 CX Amsterdam, Netherlands
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Plesmanlaan 125, 1066 CX Amsterdam, Netherlands
| | - Jurjan Aman
- Department of Pulmonary Medicine, Amsterdam UMC, location VUMC, De Boelelaan 1117, 1081 HV Amsterdam, Netherlands
| | - Lynn Mes
- Department of Rheumatology and Clinical Immunology, Amsterdam UMC, Amsterdam Rheumatology and Immunology Center, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam Infection and Immunity Institute, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Infection and Immunity Institute, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands
| | - Maurice Steenhuis
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory Academic Medical Centre, Plesmanlaan 125, 1066 CX Amsterdam, Netherlands
| | - Guillermo R Griffith
- Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam Cardiovascular Sciences, Amsterdam Infection and Immunity, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands
| | - Peter I Bonta
- Department of Pulmonology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands
| | - Philip J M Brouwer
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Infection and Immunity Institute, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands
| | - Tom G Caniels
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Infection and Immunity Institute, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands
| | - Karlijn van der Straten
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Infection and Immunity Institute, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands
- Department of Internal Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Infection and Immunity Institute, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands
| | - Korneliusz Golebski
- Department of Respiratory Medicine, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands
| | - René E Jonkers
- Department of Pulmonology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands
| | - Mads D Larsen
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Plesmanlaan 125, 1066 CX Amsterdam, Netherlands
| | - Federica Linty
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Plesmanlaan 125, 1066 CX Amsterdam, Netherlands
| | - Jan Nouta
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, 2333 AZ Leiden, Netherlands
| | - Cindy P A A van Roomen
- Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam Cardiovascular Sciences, Amsterdam Infection and Immunity, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands
| | - Frank E H P van Baarle
- Department of Intensive Care Medicine, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands
| | - Cornelis M van Drunen
- Department of Otorhinolaryngology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands
| | - Gertjan Wolbink
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory Academic Medical Centre, Plesmanlaan 125, 1066 CX Amsterdam, Netherlands
- Department of Rheumatology, Amsterdam Rheumatology and Immunology Center, Reade, Admiraal Helfrichstraat 1, 1056 AA Amsterdam, Netherlands
| | - Alexander P J Vlaar
- Department of Intensive Care Medicine, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands
| | - Godelieve J de Bree
- Department of Internal Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Infection and Immunity Institute, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands
| | - Rogier W Sanders
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Infection and Immunity Institute, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands
- Weill Medical College of Cornell University, 1300 York Avenue, New York, NY 10021, USA
| | - Lisa Willemsen
- Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam Cardiovascular Sciences, Amsterdam Infection and Immunity, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands
| | - Annette E Neele
- Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam Cardiovascular Sciences, Amsterdam Infection and Immunity, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands
| | - Diederik van de Beek
- Departments of Neurology and Neuroscience, University of Amsterdam, Meibergdreef, Amsterdam UMC, Amsterdam, Netherlands
| | - Theo Rispens
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory Academic Medical Centre, Plesmanlaan 125, 1066 CX Amsterdam, Netherlands
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, 2333 AZ Leiden, Netherlands
| | - Harm Jan Bogaard
- Department of Pulmonary Medicine, Amsterdam UMC, location VUMC, De Boelelaan 1117, 1081 HV Amsterdam, Netherlands
| | - Marit J van Gils
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Infection and Immunity Institute, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands
| | - Gestur Vidarsson
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Plesmanlaan 125, 1066 CX Amsterdam, Netherlands
| | - Menno de Winther
- Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam Cardiovascular Sciences, Amsterdam Infection and Immunity, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands.
| | - Jeroen den Dunnen
- Department of Rheumatology and Clinical Immunology, Amsterdam UMC, Amsterdam Rheumatology and Immunology Center, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands.
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam Infection and Immunity Institute, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands
| |
Collapse
|
20
|
Amado-Azevedo J, van Stalborch AMD, Valent ET, Nawaz K, van Bezu J, Eringa EC, Hoevenaars FPM, De Cuyper IM, Hordijk PL, van Hinsbergh VWM, van Nieuw Amerongen GP, Aman J, Margadant C. Depletion of Arg/Abl2 improves endothelial cell adhesion and prevents vascular leak during inflammation. Angiogenesis 2021; 24:677-693. [PMID: 33770321 PMCID: PMC7996118 DOI: 10.1007/s10456-021-09781-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 03/06/2021] [Indexed: 02/06/2023]
Abstract
Endothelial barrier disruption and vascular leak importantly contribute to organ dysfunction and mortality during inflammatory conditions like sepsis and acute respiratory distress syndrome. We identified the kinase Arg/Abl2 as a mediator of endothelial barrier disruption, but the role of Arg in endothelial monolayer regulation and its relevance in vivo remain poorly understood. Here we show that depletion of Arg in endothelial cells results in the activation of both RhoA and Rac1, increased cell spreading and elongation, redistribution of integrin-dependent cell-matrix adhesions to the cell periphery, and improved adhesion to the extracellular matrix. We further show that Arg is activated in the endothelium during inflammation, both in murine lungs exposed to barrier-disruptive agents, and in pulmonary microvessels of septic patients. Importantly, Arg-depleted endothelial cells were less sensitive to barrier-disruptive agents. Despite the formation of F-actin stress fibers and myosin light chain phosphorylation, Arg depletion diminished adherens junction disruption and intercellular gap formation, by reducing the disassembly of cell-matrix adhesions and cell retraction. In vivo, genetic deletion of Arg diminished vascular leak in the skin and lungs, in the presence of a normal immune response. Together, our data indicate that Arg is a central and non-redundant regulator of endothelial barrier integrity, which contributes to cell retraction and gap formation by increasing the dynamics of adherens junctions and cell-matrix adhesions in a Rho GTPase-dependent fashion. Therapeutic inhibition of Arg may provide a suitable strategy for the treatment of a variety of clinical conditions characterized by vascular leak.
Collapse
Affiliation(s)
- Joana Amado-Azevedo
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | | | - Erik T Valent
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Kalim Nawaz
- Sanquin Research, Amsterdam, The Netherlands
| | - Jan van Bezu
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Etto C Eringa
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Femke P M Hoevenaars
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | | | - Peter L Hordijk
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Victor W M van Hinsbergh
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Geerten P van Nieuw Amerongen
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Jurjan Aman
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Amsterdam, The Netherlands. .,Department of Pulmonology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Amsterdam, The Netherlands.
| | - Coert Margadant
- Angiogenesis Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
21
|
Kleinveld DJB, Botros L, Maas MAW, Kers J, Aman J, Hollmann MW, Juffermans NP. Bosutinib reduces endothelial permeability and organ failure in a rat polytrauma transfusion model. Br J Anaesth 2021; 126:958-966. [PMID: 33685634 PMCID: PMC8258973 DOI: 10.1016/j.bja.2021.01.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 01/17/2021] [Accepted: 01/17/2021] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND Trauma-induced shock is associated with endothelial dysfunction. We examined whether the tyrosine kinase inhibitor bosutinib as an adjunct therapy to a balanced blood component resuscitation strategy reduces trauma-induced endothelial permeability, thereby improving shock reversal and limiting transfusion requirements and organ failure in a rat polytrauma transfusion model. METHODS Male Sprague-Dawley rats (n=13 per group) were traumatised and exsanguinated until a MAP of 40 mm Hg was reached, then randomised to two groups: red blood cells, plasma and platelets in a 1:1:1 ratio with either bosutinib or vehicle. Controls were randomised to sham (median laparotomy, no trauma) with bosutinib or vehicle. Organs were harvested for histology and wet/dry (W/D) weight ratio. RESULTS Traumatic injury resulted in shock, with higher lactate levels compared with controls. In trauma-induced shock, the resuscitation volume needed to obtain a MAP of 60 mm Hg was lower in bosutinib-treated animals (2.8 [2.7-3.2] ml kg-1) compared with vehicle (6.1 [5.1-7.2] ml kg-1, P<0.001). Lactate levels in the bosutinib group were 2.9 [1.7-4.8] mM compared with 6.2 [3.1-14.1] mM in the vehicle group (P=0.06). Bosutinib compared with vehicle reduced lung vascular leakage (W/D ratio of 5.1 [4.6-5.3] vs 5.7 [5.4-6.0] (P=0.046) and lung injury scores (P=0.027). CONCLUSIONS Bosutinib as an adjunct therapy to a balanced transfusion strategy reduced resuscitation volume, improved shock reversal, and reduced vascular leak and organ injury in a rat polytrauma model.
Collapse
Affiliation(s)
- Derek J B Kleinveld
- Department of Intensive Care Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands; Laboratory of Experimental Intensive Care and Anesthesiology, Department of Trauma Surgery, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands; Department of Trauma Surgery, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.
| | - Liza Botros
- Department of Pulmonary Diseases, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands; Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - M Adrie W Maas
- Department of Intensive Care Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands; Laboratory of Experimental Intensive Care and Anesthesiology, Department of Trauma Surgery, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Jesper Kers
- Department of Pathology, Amsterdam Infection & Immunity Institute, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands; Department of Pathology, Leiden University Medical Center, University of Leiden, Leiden, The Netherlands; Van't Hoff Institute for Molecular Sciences (HIMS), University of Amsterdam, Amsterdam, The Netherlands; Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Jurjan Aman
- Department of Pulmonary Diseases, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands; Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Markus W Hollmann
- Laboratory of Experimental Intensive Care and Anesthesiology, Department of Trauma Surgery, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands; Department of Anesthesiology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Nicole P Juffermans
- Laboratory of Experimental Intensive Care and Anesthesiology, Department of Trauma Surgery, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands; Department of Intensive Care Medicine, Onze Lieve Vrouwe Gasthuis, Amsterdam, The Netherlands
| |
Collapse
|
22
|
Uddin MA, Barabutis N. P53 in the impaired lungs. DNA Repair (Amst) 2020; 95:102952. [PMID: 32846356 PMCID: PMC7437512 DOI: 10.1016/j.dnarep.2020.102952] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 08/13/2020] [Indexed: 12/12/2022]
Abstract
Our laboratory is focused on investigating the supportive role of P53 towards the maintenance of lung homeostasis. Acute lung injury, acute respiratory distress syndrome, chronic obstructive pulmonary disease, pulmonary fibrosis, bronchial asthma, pulmonary arterial hypertension, pneumonia and tuberculosis are respiratory pathologies, associated with dysfunctions of this endothelium defender (P53). Herein we review the evolving role of P53 towards the aforementioned inflammatory disorders, to potentially reveal new therapeutic possibilities in pulmonary disease.
Collapse
Affiliation(s)
- Mohammad A Uddin
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana 71201, USA
| | - Nektarios Barabutis
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana 71201, USA.
| |
Collapse
|
23
|
Duong CN, Vestweber D. Mechanisms Ensuring Endothelial Junction Integrity Beyond VE-Cadherin. Front Physiol 2020; 11:519. [PMID: 32670077 PMCID: PMC7326147 DOI: 10.3389/fphys.2020.00519] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 04/27/2020] [Indexed: 12/30/2022] Open
Abstract
Endothelial junctions provide blood and lymph vessel integrity and are essential for the formation of a vascular system. They control the extravasation of solutes, leukocytes and metastatic cells from blood vessels and the uptake of fluid and leukocytes into the lymphatic vascular system. A multitude of adhesion molecules mediate and control the integrity and permeability of endothelial junctions. VE-cadherin is arguably the most important adhesion molecule for the formation of vascular structures, and the stability of their junctions. Interestingly, despite this prominence, its elimination from junctions in the adult organism has different consequences in the vasculature of different organs, both for blood and lymph vessels. In addition, even in tissues where the lack of VE-cadherin leads to strong plasma leaks from venules, the physical integrity of endothelial junctions is preserved. Obviously, other adhesion molecules can compensate for a loss of VE-cadherin and this review will discuss which other adhesive mechanisms contribute to the stability and regulation of endothelial junctions and cooperate with VE-cadherin in intact vessels. In addition to adhesion molecules, endothelial receptors will be discussed, which stimulate signaling processes that provide junction stability by modulating the actomyosin system, which reinforces tension of circumferential actin and dampens pulling forces of radial stress fibers. Finally, we will highlight most recent reports about the formation and control of the specialized button-like junctions of initial lymphatics, which represent the entry sites for fluid and cells into the lymphatic vascular system.
Collapse
Affiliation(s)
| | - Dietmar Vestweber
- Department of Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| |
Collapse
|