1
|
Dikalova A, Ao M, Lantier L, Gutor S, Dikalov S. Depletion of Mitochondrial Cyclophilin D in Endothelial and Smooth Muscle Cells Attenuates Vascular Dysfunction and Hypertension. FUNCTION 2025; 6:zqaf006. [PMID: 39919759 PMCID: PMC11931617 DOI: 10.1093/function/zqaf006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/28/2025] [Accepted: 02/04/2025] [Indexed: 02/09/2025] Open
Abstract
Hypertension is a major risk factor of cardiovascular disease affecting nearly half of adult population. It is associated with mitochondrial dysfunction and understanding these mechanisms is important to develop new therapies. Cyclophilin D (CypD) promotes mitochondrial swelling and dysfunction. The objective of this study is to test if CypD depletion attenuates vascular dysfunction and hypertension using endothelial and smooth muscle-specific CypD knockout mice in angiotensin II model of vascular dysfunction and hypertension. Our results show that depletion of endothelial CypD prevents angiotensin II-induced impairment of endothelial-dependent vasorelaxation, preserves endothelial nitric oxide and mitochondrial respiration, attenuates hypertension, vascular oxidative stress and vascular metabolic glycolytic-switch. Depletion of smooth muscle CypD slightly reduces angiotensin II-induced hypertension, protects vascular nitric oxide and vasorelaxation, decreases vascular superoxide, diminishes angiotensin II-induced vascular glycolysis, hypertrophy and fibrosis. These data suggest "metabolic" and "redox" crosstalk between endothelial and smooth muscle cells. Endothelial CypD depletion reduces not only endothelial glycolysis but also attenuates smooth muscle cell glycolytic switch. Smooth muscle CypD depletion reduced not only smooth muscle glycolysis, but it also attenuated endothelial glycolysis. Vascular oxidative stress was inhibited both in EcCypDKO and SmcCypDKO mice, therefore, cell-specific CypD depletion had "global" antioxidant effect in vasculature. Our results support a novel function of mitochondrial CypD in regulation of superoxide and metabolism in vascular smooth muscle and endothelial cells which affect endothelial barrier and smooth muscle vascular functions. We suggest that blocking vascular CypD reduces vascular oxidative stress, improves vascular metabolism and vascular function which may be beneficial in cardiovascular disease.
Collapse
Affiliation(s)
- Anna Dikalova
- Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mingfang Ao
- Vanderbilt University Medical Center, Nashville, TN, USA
| | | | | | - Sergey Dikalov
- Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
2
|
Xie B, Xu S, Sivasankar S. Outside-in engineering of cadherin endocytosis using a conformation strengthening antibody. Nat Commun 2025; 16:1157. [PMID: 39881179 PMCID: PMC11779849 DOI: 10.1038/s41467-025-56478-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 01/15/2025] [Indexed: 01/31/2025] Open
Abstract
P-cadherin, a crucial cell-cell adhesion protein which is overexpressed in numerous malignant cancers, is a popular target for drug delivery antibodies. However, molecular guidelines for engineering antibodies that can be internalized upon binding to P-cadherin are unknown. Here, we use a combination of biophysical, biochemical, and cell biological methods to demonstrate that trapping the P-cadherin extracellular region in an X-dimer adhesive conformation triggers cadherin endocytosis via an outside-in signaling mechanism. We show that the anti-cancer drug delivery monoclonal antibody CQY684, traps P-cadherin in an X-dimer conformation and strengthens this adhesive structure. Formation of stable X-dimers results in the phosphorylation of p120-catenin, a suppressor of cadherin endocytosis. This triggers the dissociation of p120-catenin from the X-dimer cytoplasmic region, which increases P-cadherin turnover and targets the cadherin-antibody complex to the lysosome. Our results establish an outside-in signaling mechanism that provides fundamental insights into how cells regulate adhesion and that can be exploited by anti-cadherin antibodies for intracellular drug delivery.
Collapse
Affiliation(s)
- Bin Xie
- Biophysics Graduate Group, University of California, Davis, CA, USA
| | - Shipeng Xu
- Department of Biomedical Engineering, University of California, Davis, CA, USA
| | - Sanjeevi Sivasankar
- Biophysics Graduate Group, University of California, Davis, CA, USA.
- Department of Biomedical Engineering, University of California, Davis, CA, USA.
| |
Collapse
|
3
|
Onyeogaziri FC, Smith R, Arce M, Huang H, Erzar I, Rorsman C, Malinverno M, Orsenigo F, Sundell V, Fernando D, Daniel G, Niemelä M, Laakso A, Jahromi BR, Olsson AK, Magnusson PU. Pharmacological blocking of neutrophil extracellular traps attenuates immunothrombosis and neuroinflammation in cerebral cavernous malformation. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1549-1567. [PMID: 39632986 PMCID: PMC11634782 DOI: 10.1038/s44161-024-00577-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 10/29/2024] [Indexed: 12/07/2024]
Abstract
Cerebral cavernous malformation (CCM) is a neurovascular disease with symptoms such as strokes, hemorrhages and neurological deficits. With surgery being the only treatment strategy, understanding the molecular mechanisms of CCM is crucial in finding alternative therapeutic options for CCM. Neutrophil extracellular traps (NETs) were recently reported in CCM, and NETs were shown to have positive or negative effects in different disease contexts. In this study, we investigated the roles of NETs in CCM by pharmacologically inhibiting NET formation using Cl-amidine (a peptidyl arginine deiminase inhibitor). We show here that Cl-amidine treatment reduced lesion burden, coagulation and endothelial-to-mesenchymal transition. Furthermore, NETs promoted the activation of microglia and fibroblasts, leading to increased neuroinflammation and a chronic wound microenvironment in CCM. The inhibition of NET formation caused endothelial quiescence and promoted a healthier microenvironment. Our study suggests the inhibition of NETs as a potential therapeutic strategy in CCM.
Collapse
Affiliation(s)
- Favour C Onyeogaziri
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Ross Smith
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Maximiliano Arce
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Hua Huang
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Iza Erzar
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Charlotte Rorsman
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Matteo Malinverno
- Vascular Biology Unit, The FIRC Institute of Molecular Oncology Foundation, Milan, Italy
| | - Fabrizio Orsenigo
- Vascular Biology Unit, The FIRC Institute of Molecular Oncology Foundation, Milan, Italy
| | - Veronica Sundell
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Dinesh Fernando
- Department of Biomaterials and Technology/Wood Science, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Geoffrey Daniel
- Department of Biomaterials and Technology/Wood Science, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Mika Niemelä
- Department of Neurosurgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Aki Laakso
- Department of Neurosurgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Behnam Rezai Jahromi
- Department of Neurosurgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Anna-Karin Olsson
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Peetra U Magnusson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
4
|
Llaó-Cid C, Peguera B, Kobialka P, Decker L, Vogenstahl J, Alivodej N, Srivastava S, Jin J, Kirchmaier BC, Milla C, Schlierbach H, Schänzer A, Acker T, Segarra M, Acker-Palmer A. Vascular FLRT2 regulates venous-mediated angiogenic expansion and CNS barriergenesis. Nat Commun 2024; 15:10372. [PMID: 39609404 PMCID: PMC11604978 DOI: 10.1038/s41467-024-54570-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 11/12/2024] [Indexed: 11/30/2024] Open
Abstract
Veins have emerged as the origin of all other endothelial cell subtypes needed to expand vascular networks during developmental and pathological neoangiogenesis. Here, we uncover the role of the angioneurin Fibronectin Leucine Rich Transmembrane protein (FLRT) 2 in central nervous system (CNS) vascular development in the mouse. Early postnatal FLRT2 deletion reveals specific defects in retinal veins, impacting endothelial cell proliferation, sprouting and polarity that result in reduced tip cells at the vascular front. FLRT2 interacts with VE-cadherin and together with the endocytic adaptor protein Numb contribute to the modulation of adherens junction morphology in both retina and cerebral cortex in vivo. Utilizing expansion microscopy, we visualize the altered dynamic distribution of VE-cadherin in tissue of FLRT2 endothelial mutants. Additionally, FLRT2 in cortical vessels regulates the crosstalk between adherens and tight junctions, influencing blood-brain barrier development. Our findings position FLRT2 as a vein-specific regulator of CNS vascular development.
Collapse
Affiliation(s)
- C Llaó-Cid
- Buchmann Institute for Molecular Life Sciences (BMLS), Institute of Cell Biology and Neuroscience, Goethe University Frankfurt, Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany
| | - B Peguera
- Buchmann Institute for Molecular Life Sciences (BMLS), Institute of Cell Biology and Neuroscience, Goethe University Frankfurt, Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany
| | - P Kobialka
- Buchmann Institute for Molecular Life Sciences (BMLS), Institute of Cell Biology and Neuroscience, Goethe University Frankfurt, Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany
| | - L Decker
- Buchmann Institute for Molecular Life Sciences (BMLS), Institute of Cell Biology and Neuroscience, Goethe University Frankfurt, Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany
| | - J Vogenstahl
- Buchmann Institute for Molecular Life Sciences (BMLS), Institute of Cell Biology and Neuroscience, Goethe University Frankfurt, Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany
- Max Planck Institute for Brain Research, Max-von-Laue-Str. 4, 60438, Frankfurt am Main, Germany
| | - N Alivodej
- Buchmann Institute for Molecular Life Sciences (BMLS), Institute of Cell Biology and Neuroscience, Goethe University Frankfurt, Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany
- Max Planck Institute for Brain Research, Max-von-Laue-Str. 4, 60438, Frankfurt am Main, Germany
| | - S Srivastava
- Buchmann Institute for Molecular Life Sciences (BMLS), Institute of Cell Biology and Neuroscience, Goethe University Frankfurt, Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany
| | - J Jin
- Buchmann Institute for Molecular Life Sciences (BMLS), Institute of Cell Biology and Neuroscience, Goethe University Frankfurt, Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany
| | - B C Kirchmaier
- Buchmann Institute for Molecular Life Sciences (BMLS), Institute of Cell Biology and Neuroscience, Goethe University Frankfurt, Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany
| | - C Milla
- Buchmann Institute for Molecular Life Sciences (BMLS), Institute of Cell Biology and Neuroscience, Goethe University Frankfurt, Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany
| | - H Schlierbach
- Institute of Neuropathology, Justus Liebig University Giessen, D-35392, Giessen, Germany
| | - A Schänzer
- Institute of Neuropathology, Justus Liebig University Giessen, D-35392, Giessen, Germany
| | - T Acker
- Institute of Neuropathology, Justus Liebig University Giessen, D-35392, Giessen, Germany
| | - M Segarra
- Buchmann Institute for Molecular Life Sciences (BMLS), Institute of Cell Biology and Neuroscience, Goethe University Frankfurt, Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany.
- Cardio-Pulmonary Institute (CPI), Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany.
| | - A Acker-Palmer
- Buchmann Institute for Molecular Life Sciences (BMLS), Institute of Cell Biology and Neuroscience, Goethe University Frankfurt, Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany.
- Max Planck Institute for Brain Research, Max-von-Laue-Str. 4, 60438, Frankfurt am Main, Germany.
- Cardio-Pulmonary Institute (CPI), Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany.
| |
Collapse
|
5
|
Stammer P, Terhorst I, Guo J, Ibrahim A, Oberhuber A, Eierhoff T. VE-cadherin shedding in vitro and in patients with aortic aneurysm and dissection. Sci Rep 2024; 14:26743. [PMID: 39501015 PMCID: PMC11538497 DOI: 10.1038/s41598-024-77940-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024] Open
Abstract
VE-cadherin (VEC) is a major endothelial adhesion protein, which controls vascular homeostasis. During vascular diseases, VEC can be shed from the endothelial surface by proteases like ADAM10/17, which cleave the extracellular domain of VEC in response to inflammatory cytokines like TNF-α. The resulting, soluble fragments (sVEC) are discussed as a potential marker for endothelial barrier breakdown. However, its pathologic role or its potential as a specific biomarker for aortic diseases is yet unknown. Here we investigated the specificity and linkage of sVEC production with ADAM10/17 and TNF-α, both in vitro and in patients with aortic aneurysms and dissections, comparing the findings with those from patients with carotid stenosis and varicosis. Thereby, the baseline levels of sVEC, TNF-α, ADAM10 and Albumin was measured in clinical plasma samples and cell culture supernatants of human aortic endothelial cells (HAOEC) treated with TNF-α or ADAM10/17 inhibitors. The integrity of HAOEC monolayers was tested by permeability assays using Alexa488-conjugated dextran (10 kDa). Peripheral EDTA plasma samples taken preoperatively from patients ≥ 18 years of age that were diagnosed for aortic dissection (n = 29), aortic aneurysm (n = 76), carotid stenosis (n = 29) and varicose veins (n = 24) were included. In vitro shedding of VEC was induced by TNF-α and depends on ADAM10/17, which led to altered endothelial permeability. Absolute plasma sVEC levels in patients with aortic dissection (3016 ± 1008 ng/mL) and aneurysm (3288 ± 1376 ng/mL) were not statistically significantly different from patients with carotid stenosis (3013 ± 687.6 ng/mL) and varicose veins (3313 ± 1337 ng/mL). Plasma sVEC levels correlated positively with plasma TNF-α (r = 0.5586, p < 0.0001) and ADAM10 (r = 0.7003, p < 0.0001) levels with the highest degree of correlation between ADAM10 and sVEC for chronic aortic dissection (r = 0.7890, p = 0.0013), reflecting TNF-α and ADAM10 dependency of VEC shedding. In summary, VEC shedding and (plasma) sVEC levels are influenced by TNF-α and ADAM10/17 and could play a relevant role in the specific pathophysiological context of aortic diseases.
Collapse
Affiliation(s)
- Paul Stammer
- Clinic for Vascular and Endovascular Surgery, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Inka Terhorst
- Clinic for Vascular and Endovascular Surgery, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Jiangang Guo
- Clinic for Vascular and Endovascular Surgery, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
- Department of Endovascular and Vascular Surgery, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Abdulhakim Ibrahim
- Clinic for Vascular and Endovascular Surgery, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Alexander Oberhuber
- Clinic for Vascular and Endovascular Surgery, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Thorsten Eierhoff
- Clinic for Vascular and Endovascular Surgery, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany.
| |
Collapse
|
6
|
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
|
7
|
Wu L, Xiao J, Yi D, Ding H, Wang R, Duan Z, Liu Z, Shi X, Shen M, Sang J. Cytosolic Cadherin 4 promotes angiogenesis and metastasis in papillary thyroid cancer by suppressing the ubiquitination/degradation of β-catenin. J Transl Med 2024; 22:201. [PMID: 38402159 PMCID: PMC10894493 DOI: 10.1186/s12967-024-05012-1] [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: 12/24/2023] [Accepted: 02/21/2024] [Indexed: 02/26/2024] Open
Abstract
BACKGROUND Although the long-term prognosis of papillary thyroid cancer (PTC) is favorable, distant metastasis significantly compromises the prognosis and quality of life for patients with PTC. The Cadherin family plays a pivotal role in tumor metastasis; however, the involvement of Cadherin 4 (CDH4) in the metastatic cascade remains elusive. METHODS The expression and subcellular localization of CDH4 were determined through immunohistochemistry, immunofluorescence, and western blot analyses. The impact of CDH4 on cell migration, invasion, angiogenesis, and metastasis was assessed using transwell assays, tube formation assays, and animal experiments. Immunoprecipitation assay and mass spectrometry were employed to examine protein associations. The influence of CDH4 on the subcellular expression of β-catenin and active β-catenin was investigated via western blotting and immunofluorescence. Protein stability and ubiquitination assay were employed to verify the impact of CDH4 on β-catenin degradation. Rescue experiments were performed to ensure the significance of CDH4 in regulating nuclear β-catenin signaling. RESULTS CDH4 was found to be significantly overexpressed in PTC tissues and predominantly localized in the cytoplasm. Furthermore, the overexpression of CDH4 in tumor tissues is associated with lymph node metastasis in PTC patients. Cytosolic CDH4 promoted the migration, invasion, and lung metastasis of PTC cells and stimulated the angiogenesis and tumorigenesis of PTC; however, this effect could be reversed by Tegavivint, an antagonist of β-catenin. Mechanistically, cytosolic CDH4 disrupted the interaction between β-catenin and β-TrCP1, consequently impeding the ubiquitination process of β-catenin and activating the nuclear β-catenin signaling. CONCLUSIONS CDH4 induces PTC angiogenesis and metastasis via the inhibition of β-TrCP1-dependent ubiquitination of β-Catenin.
Collapse
Affiliation(s)
- Luyao Wu
- Division of Thyroid Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Jian Xiao
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Dandan Yi
- Division of Thyroid Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Haoran Ding
- Division of Thyroid Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Ru Wang
- Division of Thyroid Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Zehua Duan
- Division of Thyroid Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Zhijian Liu
- Division of Thyroid Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Xianbiao Shi
- Division of Thyroid Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Meiping Shen
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Jianfeng Sang
- Division of Thyroid Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China.
| |
Collapse
|
8
|
Balashova OA, Panoutsopoulos AA, Visina O, Selhub J, Knoepfler PS, Borodinsky LN. Noncanonical function of folate through folate receptor 1 during neural tube formation. Nat Commun 2024; 15:1642. [PMID: 38388461 PMCID: PMC10883926 DOI: 10.1038/s41467-024-45775-1] [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: 11/29/2022] [Accepted: 02/02/2024] [Indexed: 02/24/2024] Open
Abstract
Folate supplementation reduces the occurrence of neural tube defects (NTDs), birth defects consisting in the failure of the neural tube to form and close. The mechanisms underlying NTDs and their prevention by folate remain unclear. Here we show that folate receptor 1 (FOLR1) is necessary for the formation of neural tube-like structures in human-cell derived neural organoids. FOLR1 knockdown in neural organoids and in Xenopus laevis embryos leads to NTDs that are rescued by pteroate, a folate precursor that is unable to participate in metabolism. We demonstrate that FOLR1 interacts with and opposes the function of CD2-associated protein, molecule essential for apical endocytosis and turnover of C-cadherin in neural plate cells. In addition, folates increase Ca2+ transient frequency, suggesting that folate and FOLR1 signal intracellularly to regulate neural plate folding. This study identifies a mechanism of action of folate distinct from its vitamin function during neural tube formation.
Collapse
Affiliation(s)
- Olga A Balashova
- Department of Physiology & Membrane Biology, Shriners Hospitals for Children Northern California, University of California Davis, School of Medicine, Sacramento, CA, 95817, USA.
| | - Alexios A Panoutsopoulos
- Department of Physiology & Membrane Biology, Shriners Hospitals for Children Northern California, University of California Davis, School of Medicine, Sacramento, CA, 95817, USA
| | - Olesya Visina
- Department of Physiology & Membrane Biology, Shriners Hospitals for Children Northern California, University of California Davis, School of Medicine, Sacramento, CA, 95817, USA
| | - Jacob Selhub
- Tufts-USDA Human Nutrition Research Center on Aging, Boston, MA, USA
| | - Paul S Knoepfler
- Department of Cell Biology & Human Anatomy, Shriners Hospitals for Children Northern California, University of California Davis, School of Medicine, Sacramento, CA, 95817, USA
| | - Laura N Borodinsky
- Department of Physiology & Membrane Biology, Shriners Hospitals for Children Northern California, University of California Davis, School of Medicine, Sacramento, CA, 95817, USA.
| |
Collapse
|
9
|
Levine AA, Liktor-Busa E, Balasubramanian S, Palomino SM, Burtman AM, Couture SA, Lipinski AA, Langlais PR, Largent-Milnes TM. Depletion of Endothelial-Derived 2-AG Reduces Blood-Endothelial Barrier Integrity via Alteration of VE-Cadherin and the Phospho-Proteome. Int J Mol Sci 2023; 25:531. [PMID: 38203706 PMCID: PMC10778805 DOI: 10.3390/ijms25010531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 12/21/2023] [Accepted: 12/26/2023] [Indexed: 01/12/2024] Open
Abstract
Mounting evidence supports the role of the endocannabinoid system in neurophysiology, including blood-brain barrier (BBB) function. Recent work has demonstrated that activation of endocannabinoid receptors can mitigate insults to the BBB during neurological disorders like traumatic brain injury, cortical spreading depression, and stroke. As alterations to the BBB are associated with worsening clinical outcomes in these conditions, studies herein sought to examine the impact of endocannabinoid depletion on BBB integrity. Barrier integrity was investigated in vitro via bEnd.3 cell monolayers to assess endocannabinoid synthesis, barrier function, calcium influx, junctional protein expression, and proteome-wide changes. Inhibition of 2-AG synthesis using DAGLα inhibition and siRNA inhibition of DAGLα led to loss of barrier integrity via altered expression of VE-cadherin, which could be partially rescued by exogenous application of 2-AG. Moreover, the deleterious effects of DAGLα inhibition on BBB integrity showed both calcium and PKC (protein kinase C)-dependency. These data indicate that disruption of 2-AG homeostasis in brain endothelial cells, in the absence of insult, is sufficient to disrupt BBB integrity thus supporting the role of the endocannabinoid system in neurovascular disorders.
Collapse
Affiliation(s)
- Aidan A. Levine
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA; (A.A.L.); (E.L.-B.); (S.B.); (S.M.P.); (A.M.B.); (S.A.C.)
| | - Erika Liktor-Busa
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA; (A.A.L.); (E.L.-B.); (S.B.); (S.M.P.); (A.M.B.); (S.A.C.)
| | - Shreya Balasubramanian
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA; (A.A.L.); (E.L.-B.); (S.B.); (S.M.P.); (A.M.B.); (S.A.C.)
| | - Seph M. Palomino
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA; (A.A.L.); (E.L.-B.); (S.B.); (S.M.P.); (A.M.B.); (S.A.C.)
| | - Anya M. Burtman
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA; (A.A.L.); (E.L.-B.); (S.B.); (S.M.P.); (A.M.B.); (S.A.C.)
| | - Sarah A. Couture
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA; (A.A.L.); (E.L.-B.); (S.B.); (S.M.P.); (A.M.B.); (S.A.C.)
| | - Austin A. Lipinski
- Division of Endocrinology, Department of Medicine, College of Medicine, University of Arizona, Tucson, AZ 85724, USA; (A.A.L.); (P.R.L.)
| | - Paul R. Langlais
- Division of Endocrinology, Department of Medicine, College of Medicine, University of Arizona, Tucson, AZ 85724, USA; (A.A.L.); (P.R.L.)
| | - Tally M. Largent-Milnes
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA; (A.A.L.); (E.L.-B.); (S.B.); (S.M.P.); (A.M.B.); (S.A.C.)
| |
Collapse
|
10
|
Miyazaki T. Calpain and Cardiometabolic Diseases. Int J Mol Sci 2023; 24:16782. [PMID: 38069105 PMCID: PMC10705917 DOI: 10.3390/ijms242316782] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/20/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
Calpain is defined as a member of the superfamily of cysteine proteases possessing the CysPC motif within the gene. Calpain-1 and -2, which are categorized as conventional isozymes, execute limited proteolysis in a calcium-dependent fashion. Accordingly, the calpain system participates in physiological and pathological phenomena, including cell migration, apoptosis, and synaptic plasticity. Recent investigations have unveiled the contributions of both conventional and unconventional calpains to the pathogenesis of cardiometabolic disorders. In the context of atherosclerosis, overactivation of conventional calpain attenuates the barrier function of vascular endothelial cells and decreases the immunosuppressive effects attributed to lymphatic endothelial cells. In addition, calpain-6 induces aberrant mRNA splicing in macrophages, conferring atheroprone properties. In terms of diabetes, polymorphisms of the calpain-10 gene can modify insulin secretion and glucose disposal. Moreover, conventional calpain reportedly participates in amino acid production from vascular endothelial cells to induce alteration of amino acid composition in the liver microenvironment, thereby facilitating steatohepatitis. Such multifaceted functionality of calpain underscores its potential as a promising candidate for pharmaceutical targets for the treatment of cardiometabolic diseases. Consequently, the present review highlights the pivotal role of calpains in the complications of cardiometabolic diseases and embarks upon a characterization of calpains as molecular targets.
Collapse
Affiliation(s)
- Takuro Miyazaki
- Department of Biochemistry, Showa University School of Medicine, Tokyo 142-8555, Japan
| |
Collapse
|
11
|
Balashova OA, Panoutsopoulos AA, Visina O, Selhub J, Knoepfler PS, Borodinsky LN. Non-canonical function of folate/folate receptor 1 during neural tube formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.19.549718. [PMID: 37503108 PMCID: PMC10370062 DOI: 10.1101/2023.07.19.549718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Folate supplementation reduces the occurrence of neural tube defects, one of the most common and serious birth defects, consisting in the failure of the neural tube to form and close early in pregnancy. The mechanisms underlying neural tube defects and folate action during neural tube formation remain unclear. Here we show that folate receptor 1 (FOLR1) is necessary for the formation of neural tube-like structures in human-cell derived neural organoids. Knockdown of FOLR1 in human neural organoids as well as in the Xenopus laevis in vivo model leads to neural tube defects that are rescued by pteroate, a folate precursor that binds to FOLR1 but is unable to participate in metabolic pathways. We demonstrate that FOLR1 interacts with and opposes the function of CD2-associated protein (CD2AP), a molecule that we find is essential for apical endocytosis and the spatiotemporal turnover of the cell adherens junction component C-cadherin in neural plate cells. The counteracting action of FOLR1 on these processes is mediated by regulating CD2AP protein level via a degradation-dependent mechanism. In addition, folate and pteroate increase Ca 2+ transient frequency in the neural plate in a FOLR1-dependent manner, suggesting that folate/FOLR1 signal intracellularly to regulate neural plate folding. This study identifies a mechanism of action of folate distinct from its vitamin function during neural tube formation.
Collapse
|
12
|
Yi C, Chen F, Ma R, Fu Z, Song M, Zhang Z, Chen L, Tang X, Lu P, Li B, Zhang Q, Song Q, Zhu G, Wang W, Wang Q, Wang X. Serum level of calpains product as a novel biomarker of acute lung injury following cardiopulmonary bypass. Front Cardiovasc Med 2022; 9:1000761. [PMID: 36465445 PMCID: PMC9709320 DOI: 10.3389/fcvm.2022.1000761] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 10/24/2022] [Indexed: 07/30/2023] Open
Abstract
OBJECTIVE The aim of this study was to test the hypothesis whether serum level of calpains could become a meaningful biomarker for diagnosis of acute lung injury (ALI) in clinical after cardiac surgery using cardiopulmonary bypass (CPB) technology. METHODS AND RESULTS Seventy consecutive adults underwent cardiac surgery with CPB were included in this prospective study. Based on the American-European Consensus Criteria (AECC), these patients were divided into ALI (n = 20, 28.57%) and non-ALI (n = 50, 71.43%) groups. Serum level of calpains in terms of calpains' activity which was expressed as relative fluorescence unit (RFU) per microliter and measured at beginning of CPB (baseline), 1 h during CPB, end of CPB as well as 1, 12, and 24 h after CPB. Difference of serum level of calpains between two groups first appeared at the end of CPB and remained different at subsequent test points. Univariate and multivariate logistic regression analysis indicated that serum level of calpains 1 h after CPB was an independent predictor for postoperative ALI (OR 1.011, 95% CI 1.001, 1.021, p = 0.033) and correlated with a lower PaO2/FiO2 ratio in the first 2 days (The first day: r = -0.389, p < 0.001 and the second day: r = -0.320, p = 0.007) as well as longer mechanical ventilation time (r = 0.440, p < 0.001), intensive care unit (ICU) length of stay (LOS) (r = 0.419, p < 0.001) and hospital LOS (r = 0.297, p = 0.013). CONCLUSION Elevated serum level of calpains correlate with impaired lung function and poor clinical outcomes, indicating serum level of calpains could act as a potential biomarker for postoperative ALI following CPB in adults. CLINICAL TRIAL REGISTRATION [https://clinicaltrials.gov/show/NCT05610475], identifier [NCT05610475].
Collapse
Affiliation(s)
- Chenlong Yi
- Department of Cardiovascular Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Fangyu Chen
- Department of Cardiovascular Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
- Department of Thoracic and Cardiovascular Surgery, Dalian Medical University, Dalian, China
| | - Rongrong Ma
- Department of Anesthesiology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Zhi Fu
- Department of Cardiovascular Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Meijuan Song
- Jiangsu Provincial Key Laboratory of Geriatrics, Department of Geriatrics, The First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Zhuan Zhang
- Department of Anesthesiology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Lingdi Chen
- Department of Anesthesiology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Xing Tang
- Department of Operating Theatre, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Peng Lu
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ben Li
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qingfen Zhang
- Department of Anesthesiology, Peking University People’s Hospital, Beijing, China
| | - Qifeng Song
- Department of Cardiovascular Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
- Department of Thoracic and Cardiovascular Surgery, Dalian Medical University, Dalian, China
| | - Guangzheng Zhu
- Department of Cardiovascular Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
- Department of Thoracic and Cardiovascular Surgery, Dalian Medical University, Dalian, China
| | - Wei Wang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Qiang Wang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Xiaowei Wang
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
13
|
Maeso-Alonso L, Alonso-Olivares H, Martínez-García N, López-Ferreras L, Villoch-Fernández J, Puente-Santamaría L, Colas-Algora N, Fernández-Corona A, Lorenzo-Marcos ME, Jiménez B, Holmgren L, Wilhelm M, Millan J, Del Peso L, Claesson-Welsh L, Marques MM, Marin MC. p73 is required for vessel integrity controlling endothelial junctional dynamics through Angiomotin. Cell Mol Life Sci 2022; 79:535. [PMID: 36180740 PMCID: PMC9525397 DOI: 10.1007/s00018-022-04560-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 08/26/2022] [Accepted: 09/14/2022] [Indexed: 11/30/2022]
Abstract
Preservation of blood vessel integrity, which is critical for normal physiology and organ function, is controlled at multiple levels, including endothelial junctions. However, the mechanism that controls the adequate assembly of endothelial cell junctions is not fully defined. Here, we uncover TAp73 transcription factor as a vascular architect that orchestrates transcriptional programs involved in cell junction establishment and developmental blood vessel morphogenesis and identify Angiomotin (AMOT) as a TAp73 direct transcriptional target. Knockdown of p73 in endothelial cells not only results in decreased Angiomotin expression and localization at intercellular junctions, but also affects its downstream function regarding Yes-associated protein (YAP) cytoplasmic sequestration upon cell–cell contact. Analysis of adherens junctional morphology after p73-knockdown in human endothelial cells revealed striking alterations, particularly a sharp increase in serrated junctions and actin bundles appearing as stress fibers, both features associated with enhanced barrier permeability. In turn, stabilization of Angiomotin levels rescued those junctional defects, confirming that TAp73 controls endothelial junction dynamics, at least in part, through the regulation of Angiomotin. The observed defects in monolayer integrity were linked to hyperpermeability and reduced transendothelial electric resistance. Moreover, p73-knockout retinas showed a defective sprout morphology coupled with hemorrhages, highlighting the physiological relevance of p73 regulation in the maintenance of vessel integrity in vivo. We propose a new model in which TAp73 acts as a vascular architect integrating transcriptional programs that will impinge with Angiomotin/YAP signaling to maintain junctional dynamics and integrity, while balancing endothelial cell rearrangements in angiogenic vessels.
Collapse
Affiliation(s)
- Laura Maeso-Alonso
- Instituto de Biomedicina y Departamento de Biología Molecular, Universidad de León, 24071, León, Spain
| | - Hugo Alonso-Olivares
- Instituto de Biomedicina y Departamento de Biología Molecular, Universidad de León, 24071, León, Spain
| | - Nicole Martínez-García
- Instituto de Biomedicina y Departamento de Producción Animal, Universidad de León, 24071, León, Spain
| | - Lorena López-Ferreras
- Instituto de Biomedicina y Departamento de Biología Molecular, Universidad de León, 24071, León, Spain
| | - Javier Villoch-Fernández
- Instituto de Biomedicina y Departamento de Biología Molecular, Universidad de León, 24071, León, Spain
| | - Laura Puente-Santamaría
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Madrid, Spain
| | | | | | | | - Benilde Jiménez
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Madrid, Spain.,IdiPaz, Instituto de Investigación Sanitaria del Hospital Universitario La Paz, Madrid, Spain
| | - Lars Holmgren
- Department of Oncology-Pathology, Bioclinicum, Karolinska Institutet, 17164, Stockholm, Sweden
| | - Margareta Wilhelm
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 65, Stockholm, Sweden
| | - Jaime Millan
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Madrid, Spain
| | - Luis Del Peso
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Madrid, Spain.,IdiPaz, Instituto de Investigación Sanitaria del Hospital Universitario La Paz, Madrid, Spain
| | - Lena Claesson-Welsh
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Margarita M Marques
- Instituto de Desarrollo Ganadero y Sanidad Animal, y Departamento de Producción Animal, Universidad de León, 24071, León, Spain
| | - Maria C Marin
- Instituto de Biomedicina y Departamento de Biología Molecular, Universidad de León, 24071, León, Spain.
| |
Collapse
|
14
|
Surdel MC, Hahn BL, Anderson PN, Coburn J. Heterologous production of the adhesin LIC13411 from pathogenic Leptospira facilitates binding of non-pathogenic Leptospira in vitro and in vivo. Front Cell Infect Microbiol 2022; 12:917963. [PMID: 35937702 PMCID: PMC9354625 DOI: 10.3389/fcimb.2022.917963] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/27/2022] [Indexed: 01/19/2023] Open
Abstract
Leptospirosis is an important cause of morbidity and mortality worldwide. Disease severity ranges from asymptomatic colonization to widespread hemorrhage and multiorgan dysfunction. The causative agents, Leptospira spp., are zoonotic Gram-negative spirochetes. One important step in pathogenesis is binding of bacterial adhesins to host components. Previously our laboratory identified two L. interrogans candidate adhesins, LIC11574 and LIC13411, that bind to VE-cadherin in vitro. In the current study, we demonstrate the ability of two strains of pathogenic L. interrogans to disrupt the localization of VE-cadherin, a protein important to maintaining inter-endothelial junctions. Purified MBP-LIC11574 and MBP-LIC13411 bind human dermal microvascular endothelial cells in a pattern reminiscent of VE-cadherin, but do not disrupt VE-cadherin localization. Genes encoding the candidate adhesins from pathogenic Leptospira were cloned in an overexpression vector and introduced into non-pathogenic L. biflexa, creating gain-of-function strains producing LIC11574 or LIC13411. Protein production and localization to the outer membrane were confirmed by Triton X-114 fractionation. Although these strains do not disrupt VE-cadherin localization, production of LIC13411 increases binding of non-pathogenic Leptospira to human endothelial cells and specifically to VE-cadherin. In a short-term murine model of infection, LIC13411 production led to increased burdens of the non-pathogen in the lung, liver, kidney, and bladder. These data confirm the role of LIC13411 as an adhesin in Leptospira spp. and implicate it in dissemination to multiple organs. Importantly, anti-adhesin therapy has been shown to have many benefits over classical antibiotics. Taken together, this work provides novel insight into the pathogenesis of Leptospira spp. and identifies LIC13411 as a potential prophylactic and therapeutic target.
Collapse
Affiliation(s)
- Matthew C. Surdel
- Department of Medicine, Division of Infectious Diseases, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Beth L. Hahn
- Department of Medicine, Division of Infectious Diseases, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Phillip N. Anderson
- Department of Medicine, Division of Infectious Diseases, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Jenifer Coburn
- Department of Medicine, Division of Infectious Diseases, Medical College of Wisconsin, Milwaukee, WI, United States,Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, United States,*Correspondence: Jenifer Coburn,
| |
Collapse
|
15
|
Giri H, Srivastava AK, Naik UP. Apoptosis signal-regulating kinase-1 regulates thrombin-induced endothelial permeability. Vascul Pharmacol 2022; 145:107088. [DOI: 10.1016/j.vph.2022.107088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/17/2022] [Accepted: 06/29/2022] [Indexed: 11/16/2022]
|
16
|
Yang M, Li S, Huang L, Zhao R, Dai E, Jiang X, He Y, Lu J, Peng L, Liu W, Zhang Z, Jiang D, Zhang Y, Jiang Z, Yang Y, Zhao P, Zhu X, Ding X, Yang Z. CTNND1 variants cause familial exudative vitreoretinopathy through Wnt/Cadherin axis. JCI Insight 2022; 7:158428. [PMID: 35700046 PMCID: PMC9431724 DOI: 10.1172/jci.insight.158428] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 06/10/2022] [Indexed: 11/17/2022] Open
Abstract
Familial exudative vitreoretinopathy (FEVR) is a hereditary disorder that can cause vision loss. The CTNND1 gene encodes a cellular adhesion protein p120-catenin (p120), which is essential for vascularization, yet the function of p120 in postnatal physiological angiogenesis remains unclear. Here, we applied whole-exome sequencing (WES) on 140 probands of FEVR families and identified three candidate variants in the human CTNND1 gene. We performed inducible deletion of Ctnnd1 in the postnatal mouse endothelial cells (ECs) and observed typical phenotypes of FEVR. Immunofluorescence of retina flat mounts also revealed immune responses, including reactive astrogliosis and microgliosis accompanied by abnormal Vegfa expression. Using an unbiased proteomics analysis in combination with in vivo or in vitro approaches, we propose that p120 is critical for the integrity of cadherin/catenin complex, and that p120 activates Wnt signaling activity by protecting β-catenin from Gsk3β-ubiquitin-guided degradation. Treatment of CTNND1-depleted HRECs with Gsk3β inhibitors LiCl or CHIR-99021 successfully enhanced cell proliferation by preventing β-catenin from degradation. Moreover, LiCl treatment increased vessel density in Ctnnd1-deficient mouse retinas. Functional analysis also revealed that variants in CTNND1 cause FEVR by compromising the expression of adherens junctions (AJs) and Wnt signaling activity. Additionally, genetic interactions between p120 and β-catenin or α-catenin revealed by double heterozygous deletion in mice further confirmed that p120 regulates vascular development through the Wnt/Cadherin axis. Together, we propose that CTNND1 is a novel candidate gene associated with FEVR, and that variants in CTNND1 can cause FEVR through the Wnt/Cadherin axis.
Collapse
Affiliation(s)
- Mu Yang
- Prenatal Diagnosis Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Shujin Li
- Prenatal Diagnosis Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Li Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Rulian Zhao
- Prenatal Diagnosis Center, Sichuan Provincial Key Laboratory for Human Disease Gene Study, Prenatal Diagnosis Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Erkuan Dai
- Department of Ophthalmology, Shanghai Jiaotong University School of Medicine Xinhua Hospital, Chengdu, China
| | - Xiaoyan Jiang
- Center for Human Molecular Genetics, University of Electronic Science and Technology of China, Chengdu, China
| | - Yunqi He
- Center for Human Molecular Genetics, University of Electronic Science and Technology of China, Chengdu, China
| | - Jinglin Lu
- Prenatal Diagnosis Center, Sun Yat-sen University, Guangzhou, China
| | - Li Peng
- Center for Human Molecular Genetics, Sun Yat-sen University, Chengdu, China
| | - Wenjing Liu
- Center for Human Molecular Genetics, Sun Yat-sen University, Chengdu, China
| | - Zhaotian Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Dan Jiang
- The Key Laboratory for Human Disease Gene Study of Sichuan Province, Sichua, Chengdu, China
| | - Yi Zhang
- Center for Human Molecular Genetics, University of Electronic Science and Technology of China, Chengdu, China
| | - Zhilin Jiang
- Center for Human Molecular Genetics, University of Electronic Science and Technology of China, Chengdu, China
| | - Yeming Yang
- Center for Human Molecular Genetics, University of Electronic Science and Technology of China, Chengdu, China
| | - Peiquan Zhao
- Department of Ophthalmology, Shanghai Jiaotong University School of Medicine Xinhua Hospital, Chengdu, China
| | - Xianjun Zhu
- Center for Human Molecular Genetics, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaoyan Ding
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Zhenglin Yang
- Department of Medical Genetics, Sichuan Provincial Key Laboratory for Human Disease Gene Study, Prenatal Diagnosis Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
17
|
Francis CR, Kushner EJ. Trafficking in blood vessel development. Angiogenesis 2022; 25:291-305. [PMID: 35449244 PMCID: PMC9249721 DOI: 10.1007/s10456-022-09838-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 04/03/2022] [Indexed: 02/17/2023]
Abstract
Blood vessels demonstrate a multitude of complex signaling programs that work in concert to produce functional vasculature networks during development. A known, but less widely studied, area of endothelial cell regulation is vesicular trafficking, also termed sorting. After moving through the Golgi apparatus, proteins are shuttled to organelles, plugged into membranes, recycled, or degraded depending on the internal and extrinsic cues. A snapshot of these protein-sorting systems can be viewed as a trafficking signature that is not only unique to endothelial tissue, but critically important for blood vessel form and function. In this review, we will cover how vesicular trafficking impacts various aspects of angiogenesis, such as sprouting, lumen formation, vessel stabilization, and secretion, emphasizing the role of Rab GTPase family members and their various effectors.
Collapse
Affiliation(s)
- Caitlin R Francis
- Department of Biological Sciences, University of Denver, Denver, CO, 80210, USA
| | - Erich J Kushner
- Department of Biological Sciences, University of Denver, Denver, CO, 80210, USA.
| |
Collapse
|
18
|
Kertesz Z, Harrington EO, Braza J, Guarino BD, Chichger H. Agonists for Bitter Taste Receptors T2R10 and T2R38 Attenuate LPS-Induced Permeability of the Pulmonary Endothelium in vitro. Front Physiol 2022; 13:794370. [PMID: 35399266 PMCID: PMC8985831 DOI: 10.3389/fphys.2022.794370] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 02/25/2022] [Indexed: 12/12/2022] Open
Abstract
One of the hallmarks of acute respiratory distress syndrome (ARDS) is an excessive increase in pulmonary vascular permeability. In settings of ARDS, the loss of barrier integrity is mediated by cell-cell contact disassembly and actin remodelling. Studies into molecular mechanisms responsible for improving microvascular barrier function are therefore vital in the development of therapeutic targets for reducing vascular permeability seen in ARDS. Bitter taste receptors (T2Rs) belong to the superfamily of G-protein-coupled receptors found in several extraoral systems, including lung epithelial and smooth muscle cells. In the present study, we show for the first time that several T2Rs are expressed in human pulmonary arterial endothelial cells (HPAECs). Our results focus on those which are highly expressed as: T2R10, T2R14 and T2R38. Agonists for T2R10 (denatonium) and T2R38 (phenylthiourea), but not T2R14 (noscapine), significantly attenuated lipopolysaccharide (LPS)-induced permeability and VE-cadherin internalisation in HPAECs. In T2R10- or T2R38-siRNA knockdown cells, these endothelial-protective effects were abolished, indicating a direct effect of agonists in regulating barrier integrity. Our further findings indicate that T2R10 and T2R38 exert their barrier-protective function through cAMP but via Rac1-dependent and independent pathways, respectively. However, using an in vivo model of ARDS, the T2R38 agonist, phenylthiourea, was not able to protect against pulmonary edema formation. Taken together, these studies identify bitter taste sensing in the pulmonary endothelium to regulate barrier integrity in vitro through cAMP-Rac1 signalling.
Collapse
Affiliation(s)
- Zsuzsanna Kertesz
- Biomedical Research Group, Anglia Ruskin University, Cambridge, United Kingdom
| | - Elizabeth O. Harrington
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, RI, United States
- Department of Medicine, Alpert Medical School of Brown University, Providence, RI, United States
| | - Julie Braza
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, RI, United States
- Department of Medicine, Alpert Medical School of Brown University, Providence, RI, United States
| | - Brianna D. Guarino
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, RI, United States
- Department of Medicine, Alpert Medical School of Brown University, Providence, RI, United States
| | - Havovi Chichger
- Biomedical Research Group, Anglia Ruskin University, Cambridge, United Kingdom
| |
Collapse
|
19
|
Wu Y, Yu X, Wang Y, Huang Y, Tang J, Gong S, Jiang S, Xia Y, Li F, Yu B, Zhang Y, Kou J. Ruscogenin alleviates LPS-triggered pulmonary endothelial barrier dysfunction through targeting NMMHC IIA to modulate TLR4 signaling. Acta Pharm Sin B 2022; 12:1198-1212. [PMID: 35530141 PMCID: PMC9069402 DOI: 10.1016/j.apsb.2021.09.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/16/2021] [Accepted: 09/07/2021] [Indexed: 12/03/2022] Open
Abstract
Pulmonary endothelial barrier dysfunction is a hallmark of clinical pulmonary edema and contributes to the development of acute lung injury (ALI). Here we reported that ruscogenin (RUS), an effective steroidal sapogenin of Radix Ophiopogon japonicus, attenuated lipopolysaccharides (LPS)-induced pulmonary endothelial barrier disruption through mediating non-muscle myosin heavy chain IIA (NMMHC IIA)‒Toll-like receptor 4 (TLR4) interactions. By in vivo and in vitro experiments, we observed that RUS administration significantly ameliorated LPS-triggered pulmonary endothelial barrier dysfunction and ALI. Moreover, we identified that RUS directly targeted NMMHC IIA on its N-terminal and head domain by serial affinity chromatography, molecular docking, biolayer interferometry, and microscale thermophoresis analyses. Downregulation of endothelial NMMHC IIA expression in vivo and in vitro abolished the protective effect of RUS. It was also observed that NMMHC IIA was dissociated from TLR4 and then activating TLR4 downstream Src/vascular endothelial cadherin (VE-cadherin) signaling in pulmonary vascular endothelial cells after LPS treatment, which could be restored by RUS. Collectively, these findings provide pharmacological evidence showing that RUS attenuates LPS-induced pulmonary endothelial barrier dysfunction by inhibiting TLR4/Src/VE-cadherin pathway through targeting NMMHC IIA and mediating NMMHC IIA‒TLR4 interactions.
Collapse
|
20
|
To Stick or Not to Stick: Adhesions in Orofacial Clefts. BIOLOGY 2022; 11:biology11020153. [PMID: 35205020 PMCID: PMC8869391 DOI: 10.3390/biology11020153] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 11/17/2022]
Abstract
Morphogenesis requires a tight coordination between mechanical forces and biochemical signals to inform individual cellular behavior. For these developmental processes to happen correctly the organism requires precise spatial and temporal coordination of the adhesion, migration, growth, differentiation, and apoptosis of cells originating from the three key embryonic layers, namely the ectoderm, mesoderm, and endoderm. The cytoskeleton and its remodeling are essential to organize and amplify many of the signaling pathways required for proper morphogenesis. In particular, the interaction of the cell junctions with the cytoskeleton functions to amplify the behavior of individual cells into collective events that are critical for development. In this review we summarize the key morphogenic events that occur during the formation of the face and the palate, as well as the protein complexes required for cell-to-cell adhesions. We then integrate the current knowledge into a comprehensive review of how mutations in cell-to-cell adhesion genes lead to abnormal craniofacial development, with a particular focus on cleft lip with or without cleft palate.
Collapse
|
21
|
Zhu X, Yang M, Zhao P, Li S, Zhang L, Huang L, Huang Y, Fei P, Yang Y, Zhang S, Xu H, Yuan Y, Zhang X, Zhu X, Ma S, Hao F, Sundaresan P, Zhu W, Yang Z. Catenin α 1 mutations cause familial exudative vitreoretinopathy by overactivating Norrin/β-catenin signaling. J Clin Invest 2021; 131:139869. [PMID: 33497368 DOI: 10.1172/jci139869] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 01/22/2021] [Indexed: 12/19/2022] Open
Abstract
Familial exudative vitreoretinopathy (FEVR) is a severe retinal vascular disease that causes blindness. FEVR has been linked to mutations in several genes associated with inactivation of the Norrin/β-catenin signaling pathway, but these account for only approximately 50% of cases. We report that mutations in α-catenin (CTNNA1) cause FEVR by overactivating the β-catenin pathway and disrupting cell adherens junctions. We identified 3 heterozygous mutations in CTNNA1 (p.F72S, p.R376Cfs*27, and p.P893L) by exome sequencing and further demonstrated that FEVR-associated mutations led to overactivation of Norrin/β-catenin signaling as a result of impaired protein interactions within the cadherin-catenin complex. The clinical features of FEVR were reproduced in mice lacking Ctnna1 in vascular endothelial cells (ECs) or with overactivated β-catenin signaling by an EC-specific gain-of-function allele of Ctnnb1. In isolated mouse lung ECs, both CTNNA1-P893L and F72S mutants failed to rescue either the disrupted F-actin arrangement or the VE-cadherin and CTNNB1 distribution. Moreover, we discovered that compound heterozygous Ctnna1 F72S and a deletion allele could cause a similar phenotype. Furthermore, in a FEVR family, we identified a mutation of LRP5, which activates Norrin/β-catenin signaling, and the corresponding knockin mice exhibited a partial FEVR-like phenotype. Our study demonstrates that the precise regulation of β-catenin activation is critical for retinal vascular development and provides new insights into the pathogenesis of FEVR.
Collapse
Affiliation(s)
- Xianjun Zhu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.,Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China.,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Mu Yang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.,Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Peiquan Zhao
- Department of Ophthalmology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Shujin Li
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.,Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Lin Zhang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Lulin Huang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yi Huang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Ping Fei
- Department of Ophthalmology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yeming Yang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Shanshan Zhang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Huijuan Xu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Ye Yuan
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Xiang Zhang
- Department of Ophthalmology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Xiong Zhu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Shi Ma
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Fang Hao
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Periasamy Sundaresan
- Department of Genetics, Aravind Medical Research Foundation, Aravind Eye Hospital, Madurai, Tamil Nadu, India
| | - Weiquan Zhu
- Department of Molecular Medicine, School of Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Zhenglin Yang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.,Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China.,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| |
Collapse
|
22
|
Guo XW, Zhang H, Huang JQ, Wang SN, Lu Y, Cheng B, Dong SH, Wang YY, Li FS, Li YW. PIEZO1 Ion Channel Mediates Ionizing Radiation-Induced Pulmonary Endothelial Cell Ferroptosis via Ca 2+/Calpain/VE-Cadherin Signaling. Front Mol Biosci 2021; 8:725274. [PMID: 34568428 PMCID: PMC8458942 DOI: 10.3389/fmolb.2021.725274] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/20/2021] [Indexed: 12/23/2022] Open
Abstract
Pulmonary endothelial cell dysfunction plays an important role in ionizing radiation (IR)-induced lung injury. Whether pulmonary endothelial cell ferroptosis occurs after IR and what are the underlying mechanisms remain elusive. Here, we demonstrate that 15-Gy IR induced ferroptosis characterized by lethal accumulation of reactive oxygen species (ROS), lipid peroxidation, mitochondria shrinkage, and decreased glutathione peroxidase 4 (GPX4) and SLC7A11 expression in pulmonary endothelial cells. The phenomena could be mimicked by Yoda1, a specific activator of mechanosensitive calcium channel PIEZO1. PIEZO1 protein expression was upregulated by IR in vivo and in vitro. The increased PIEZO1 expression after IR was accompanied with increased calcium influx and increased calpain activity. The effects of radiation on lung endothelial cell ferroptosis was partly reversed by inhibition of PIEZO1 activity using the selective inhibitor GsMTx4 or inhibition of downstreaming Ca2+/calpain signaling using PD151746. Both IR and activation of PIEZO1 led to increased degradation of VE-cadherin, while PD151746 blocked these effects. VE-cadherin knockdown by specific siRNA causes ferroptosis-like phenomena with increased ROS and lipid peroxidation in the lung endothelial cells. Overexpression of VE-cadherin partly recused the ferroptosis caused by IR or PIEZO1 activation as supported by decreased ROS production, lipid peroxidation and mitochondria shrinkage compared to IR or PIEZO1 activation alone. In summary, our study reveals a previously unrecognized role of PIEZO1 in modulating ferroptosis, providing a new target for future mitigation of radiation-induced lung injury.
Collapse
Affiliation(s)
- Xue-Wei Guo
- The Postgraduate Training Base of Jinzhou Medical University (The PLA Rocket Force Characteristic Medical Center), Beijing, China.,Department of Anesthesiology, The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Hao Zhang
- Department of Anesthesiology, The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Jia-Qi Huang
- The Postgraduate Training Base of Jinzhou Medical University (The PLA Rocket Force Characteristic Medical Center), Beijing, China.,Department of Anesthesiology, The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Si-Nian Wang
- Department of Nuclear Radiation Injury and Monitoring, The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Yan Lu
- Department of Neurology, The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Bo Cheng
- Department of Pathology, The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Su-He Dong
- Department of Nuclear Radiation Injury and Monitoring, The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Ying-Ying Wang
- Department of Anesthesiology, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Feng-Sheng Li
- Department of Nuclear Radiation Injury and Monitoring, The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Yong-Wang Li
- Department of Anesthesiology, The PLA Rocket Force Characteristic Medical Center, Beijing, China
| |
Collapse
|
23
|
Zhang L, Ma L, Li J, Lei J, Chen J, Yu C. VE-cadherin N-glycosylation modified by N-acetylglucosaminyltransferase V regulates VE-cadherin-β-catenin interaction and monocyte adhesion. Exp Physiol 2021; 106:1869-1877. [PMID: 34117813 DOI: 10.1113/ep089617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 06/10/2021] [Indexed: 11/08/2022]
Abstract
NEW FINDINGS What is the central question of this study? Inflammation-induced monocyte adhesion is the initiator of most vascular diseases. The underlying mechanisms that mediate monocyte adhesion remain to be clarified fully. What is the main finding and its importance? N-acetylglucosaminyltransferase V (GnT-V)-mediated N-glycosylation of VE-cadherin regulates the dissociation of the VE-cadherin-β-catenin complex to modulate monocyte adhesion, but GnT-V overexpression cannot rescue monocyte adhesion induced by interleukin-1β. This study clarified the molecular mechanism of VE-cadherin in regulating the monocyte adhesion process. ABSTRACT Monocyte adhesion is a crucial step in the initial stage of atherosclerosis, and dysfunction of VE-cadherin has been reported to be involved in this process. Our group previously found that VE-cadherin and its binding protein, β-catenin, were modified by sialylation, and the levels of sialylation were decreased in pro-inflammatory cytokine-treated human umbilical vein EA.hy926 cells. In this study, we confirmed that the sugar chains of VE-cadherin were modified by N-acetylglucosaminyltransferase V (GnT-V). We showed that the levels of GnT-V and β1,6-N-acetylglucosamine on the VE-cadherin were reduced in the presence of interleukin-1β, whereas the level of monocyte transendothelial migration was increased. Moreover, the interaction between VE-cadherin and β-catenin was increased, accompanied by an increased accumulation of degradative VE-cadherin and cytoplasmic β-catenin, indicating impairment of cell-cell junctions after interleukin-1β treatment. Furthermore, GnT-V short hairpin RNA and overexpression analysis confirmed that glycosylation of VE-cadherin was modified by GnT-V in EA.hy926 cells, which contributed to the monocyte-endothelial adhesion process. Taken together, these results suggest that the function of VE-cadherin in facilitating monocyte adhesion might result from the decreasing GnT-V expression and disorder of GnT-V-catalysed N-glycosylation. Our study clarified the molecular mechanism of VE-cadherin in regulation of the monocyte adhesion process and provided new insights into the post-transcriptional modifications of VE-cadherin.
Collapse
Affiliation(s)
- Lei Zhang
- College of Pharmacy, Chongqing Medical University, Chongqing, PR China.,Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, PR China
| | - Limei Ma
- College of Pharmacy, Chongqing Medical University, Chongqing, PR China.,Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, PR China
| | - Jiajia Li
- College of Pharmacy, Chongqing Medical University, Chongqing, PR China.,Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, PR China.,Department of Pharmacy, Chongqing Hechuan District People's Hospital, Chongqing, PR China
| | - Jin Lei
- College of Pharmacy, Chongqing Medical University, Chongqing, PR China.,Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, PR China
| | - Jun Chen
- College of Pharmacy, Chongqing Medical University, Chongqing, PR China.,Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, PR China
| | - Chao Yu
- College of Pharmacy, Chongqing Medical University, Chongqing, PR China.,Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, PR China
| |
Collapse
|
24
|
Jiang L, Zhang Y, Lu D, Huang T, Yan K, Yang W, Gao J. Mechanosensitive Piezo1 channel activation promotes ventilator-induced lung injury via disruption of endothelial junctions in ARDS rats. Biochem Biophys Res Commun 2021; 556:79-86. [PMID: 33839418 DOI: 10.1016/j.bbrc.2021.03.163] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 03/29/2021] [Indexed: 12/15/2022]
Abstract
OBJECTIVE This study aimed to investigate the role of endothelial Piezo1 in mediating ventilator-induced lung injury secondary to acute respiratory distress syndrome (ARDS). METHODS Rats and lung endothelial cells (ECs) were transfected with Piezo1 shRNA (shPiezo1) and Piezo1 siRNA, respectively, to knock down Piezo1. Intratracheal instillation or incubation with lipopolysaccharide (LPS) was used to establish an ARDS model, and high tidal volume (HVT) ventilation or 20% cyclic stretch (CS) was administered to simulate a two-hit injury. Lung injury, alterations in lung endothelial barrier, disruption of adherens junctions (AJs), and Ca2+ influx were assessed. RESULTS Lung vascular hyperpermeability was further increased in ARDS rats following HVT ventilation, which was abrogated in shPiezo1-treated rats. 20% CS led to severer rupture of AJs following LPS stimulation as indicated by immunofluorescence staining. The internalization and degradation of VE-cadherin were blocked by knockdown of Piezo1. Additionally, 20% CS induced Piezo1 activation, manifesting as elevated intracellular Ca2+ concentration in LPS-treated ECs, and subsequently increased calcium-dependent calpain activity. Pharmacological inhibition of calpain or Piezo1 knockdown prevented the loss of VE-cadherin, p120-catenin, and β-catenin in ARDS rats undergoing HVT ventilation and LPS-treated ECs exposed to 20% CS. CONCLUSION Excessive mechanical stretch during ARDS induces the activation of Piezo1 channel and its downstream target, calpain, via Ca2+ influx. This results in the disassembly of endothelial AJs and further facilitates lung endothelial barrier breakdown and vascular hyperpermeability.
Collapse
Affiliation(s)
- Lulu Jiang
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Yang Zhang
- Department of Anesthesiology, Northern Jiangsu People's Hospital, Clinical Medical School, Yangzhou University, Yangzhou, 225001, China
| | - Dahao Lu
- Department of Anesthesiology, Northern Jiangsu People's Hospital, Clinical Medical School, Yangzhou University, Yangzhou, 225001, China
| | - Tianfeng Huang
- Department of Anesthesiology, Northern Jiangsu People's Hospital, Clinical Medical School, Yangzhou University, Yangzhou, 225001, China
| | - Keshi Yan
- Department of Anesthesiology, Northern Jiangsu People's Hospital, Clinical Medical School, Yangzhou University, Yangzhou, 225001, China
| | - Wenjun Yang
- Department of Anesthesiology, Northern Jiangsu People's Hospital, Clinical Medical School, Yangzhou University, Yangzhou, 225001, China
| | - Ju Gao
- Department of Anesthesiology, Northern Jiangsu People's Hospital, Clinical Medical School, Yangzhou University, Yangzhou, 225001, China.
| |
Collapse
|
25
|
Anisodamine Hydrobromide Protects Glycocalyx and Against the Lipopolysaccharide-Induced Increases in Microvascular Endothelial Layer Permeability and Nitric Oxide Production. Cardiovasc Eng Technol 2020; 12:91-100. [PMID: 32935201 DOI: 10.1007/s13239-020-00486-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 09/08/2020] [Indexed: 02/08/2023]
Abstract
PURPOSE Anisodamine hydrobromide (Ani HBr) has been used to improve the microcirculation during cardiovascular disorders and sepsis. Glycocalyx plays an important role in preserving the endothelial cell (EC) barrier permeability and nitric oxide (NO) production. We aimed to test the hypothesis that Ani HBr could protect the EC against permeability and NO production via preventing glycocalyx shedding. METHODS A human cerebral microvascular EC hCMEC/D3 injury model induced by lipopolysaccharide (LPS) was established. Ani HBr was administrated to ECs with the LPS challenge. Cell viability was performed by Cell Counting Kit-8 assay. Cell proliferation and apoptosis were detected by EdU and Hoechst 33342 staining. Apoptosis and cell cycle were also assessed by flow cytometry with annexin V staining and propidium iodide staining, respectively. Then, adherens junction integrity was evaluated basing on the immunofluorescence staining of vascular endothelial cadherin (VE-cadherin). The glycocalyx component heparan sulfate (HS) was stained in ECs. The cell permeability was evaluated by leakage of fluorescein isothiocyanate (FITC)-dextran. Cellular NO production was measured by the method of nitric acid reductase. RESULTS Ani HBr at 20 μg/mL significantly increased the viability of ECs with LPS challenge, but significantly inhibited the cell viability at 80 μg/mL, showing a bidirectional regulation of cell viability by Ani HBr. Ani HBr had not significantly change the LPS-induced EC proliferation. Ani HBr significantly reversed the induction of LPS on EC apoptosis. Ani HBr reinstated the LPS-induced glycocalyx and VE-cadherin shedding and adherens junction disruption. Ani HBr significantly alleviated LPS-induced EC layer permeability and NO production. CONCLUSION Ani HBr protects ECs against LPS-induced increase in cell barrier permeability and nitric oxide production via preserving the integrity of glycocalyx. Ani HBr is a promising drug to rescue or protect the glycocalyx.
Collapse
|
26
|
Colás-Algora N, García-Weber D, Cacho-Navas C, Barroso S, Caballero A, Ribas C, Correas I, Millán J. Compensatory increase of VE-cadherin expression through ETS1 regulates endothelial barrier function in response to TNFα. Cell Mol Life Sci 2020; 77:2125-2140. [PMID: 31396656 PMCID: PMC11105044 DOI: 10.1007/s00018-019-03260-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 07/24/2019] [Accepted: 07/29/2019] [Indexed: 02/07/2023]
Abstract
VE-cadherin plays a central role in controlling endothelial barrier function, which is transiently disrupted by proinflammatory cytokines such as tumor necrosis factor (TNFα). Here we show that human endothelial cells compensate VE-cadherin degradation in response to TNFα by inducing VE-cadherin de novo synthesis. This compensation increases adherens junction turnover but maintains surface VE-cadherin levels constant. NF-κB inhibition strongly reduced VE-cadherin expression and provoked endothelial barrier collapse. Bacterial lipopolysaccharide and TNFα upregulated the transcription factor ETS1, in vivo and in vitro, in an NF-κB dependent manner. ETS1 gene silencing specifically reduced VE-cadherin protein expression in response to TNFα and exacerbated TNFα-induced barrier disruption. We propose that TNFα induces not only the expression of genes involved in increasing permeability to small molecules and immune cells, but also a homeostatic transcriptional program in which NF-κB- and ETS1-regulated VE-cadherin expression prevents the irreversible damage of endothelial barriers.
Collapse
Affiliation(s)
| | - Diego García-Weber
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), 28049, Madrid, Spain.
- INSERM, U1016, Institut Cochin, Paris, France.
| | | | - Susana Barroso
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), 28049, Madrid, Spain
| | - Alvaro Caballero
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), 28049, Madrid, Spain
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, 28049, Madrid, Spain
- Instituto de Investigación Sanitaria La Princesa, 28006, Madrid, Spain
| | - Catalina Ribas
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), 28049, Madrid, Spain
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, 28049, Madrid, Spain
- Instituto de Investigación Sanitaria La Princesa, 28006, Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, ISCIII (CIBERCV), 28029, Madrid, Spain
| | - Isabel Correas
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), 28049, Madrid, Spain
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | - Jaime Millán
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), 28049, Madrid, Spain.
| |
Collapse
|
27
|
Fu P, Ramchandran R, Shaaya M, Huang L, Ebenezer DL, Jiang Y, Komarova Y, Vogel SM, Malik AB, Minshall RD, Du G, Tonks NK, Natarajan V. Phospholipase D2 restores endothelial barrier function by promoting PTPN14-mediated VE-cadherin dephosphorylation. J Biol Chem 2020; 295:7669-7685. [PMID: 32327488 DOI: 10.1074/jbc.ra119.011801] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 04/03/2020] [Indexed: 11/06/2022] Open
Abstract
Increased permeability of vascular lung tissues is a hallmark of acute lung injury and is often caused by edemagenic insults resulting in inflammation. Vascular endothelial (VE)-cadherin undergoes internalization in response to inflammatory stimuli and is recycled at cell adhesion junctions during endothelial barrier re-establishment. Here, we hypothesized that phospholipase D (PLD)-generated phosphatidic acid (PA) signaling regulates VE-cadherin recycling and promotes endothelial barrier recovery by dephosphorylating VE-cadherin. Genetic deletion of PLD2 impaired recovery from protease-activated receptor-1-activating peptide (PAR-1-AP)-induced lung vascular permeability and potentiated inflammation in vivo In human lung microvascular endothelial cells (HLMVECs), inhibition or deletion of PLD2, but not of PLD1, delayed endothelial barrier recovery after thrombin stimulation. Thrombin stimulation of HLMVECs increased co-localization of PLD2-generated PA and VE-cadherin at cell-cell adhesion junctions. Inhibition of PLD2 activity resulted in prolonged phosphorylation of Tyr-658 in VE-cadherin during the recovery phase 3 h post-thrombin challenge. Immunoprecipitation experiments revealed that after HLMVECs are thrombin stimulated, PLD2, VE-cadherin, and protein-tyrosine phosphatase nonreceptor type 14 (PTPN14), a PLD2-dependent protein-tyrosine phosphatase, strongly associate with each other. PTPN14 depletion delayed VE-cadherin dephosphorylation, reannealing of adherens junctions, and barrier function recovery. PLD2 inhibition attenuated PTPN14 activity and reversed PTPN14-dependent VE-cadherin dephosphorylation after thrombin stimulation. Our findings indicate that PLD2 promotes PTPN14-mediated dephosphorylation of VE-cadherin and that redistribution of VE-cadherin at adherens junctions is essential for recovery of endothelial barrier function after an edemagenic insult.
Collapse
Affiliation(s)
- Panfeng Fu
- Department of Pharmacology, University of Illinois, Chicago, Illinois.,The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | | | - Mark Shaaya
- Department of Pharmacology, University of Illinois, Chicago, Illinois
| | - Longshuang Huang
- Department of Pharmacology, University of Illinois, Chicago, Illinois
| | - David L Ebenezer
- Department of Pharmacology, University of Illinois, Chicago, Illinois
| | - Ying Jiang
- Department of Anesthesiology, University of Illinois, Chicago, Illinois
| | - Yulia Komarova
- Department of Pharmacology, University of Illinois, Chicago, Illinois
| | - Stephen M Vogel
- Department of Pharmacology, University of Illinois, Chicago, Illinois
| | - Asrar B Malik
- Department of Pharmacology, University of Illinois, Chicago, Illinois
| | - Richard D Minshall
- Department of Pharmacology, University of Illinois, Chicago, Illinois.,Department of Anesthesiology, University of Illinois, Chicago, Illinois
| | - Guangwei Du
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, Texas
| | | | - Viswanathan Natarajan
- Department of Pharmacology, University of Illinois, Chicago, Illinois .,Department of Medicine, University of Illinois, Chicago, Illinois
| |
Collapse
|
28
|
Calpain proteolytic systems counteract endothelial cell adaptation to inflammatory environments. Inflamm Regen 2020; 40:5. [PMID: 32266045 PMCID: PMC7114782 DOI: 10.1186/s41232-020-00114-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 03/23/2020] [Indexed: 02/08/2023] Open
Abstract
Vascular endothelial cells (ECs) make up the innermost surface of arteries, veins, and capillaries, separating the remaining layers of the vessel wall from circulating blood. Under non-inflammatory conditions, ECs are quiescent and form a robust barrier structure; however, exposure to inflammatory stimuli induces changes in the expression of EC proteins that control transcellular permeability and facilitate angiogenic tube formation. Increasing evidence suggests that dysfunction in intracellular proteolytic systems disturbs EC adaptation to the inflammatory environment, leading to vascular disorders such as atherosclerosis and pathological angiogenesis. Recent work has highlighted the contribution of the calpain–calpastatin stress-responsive intracellular proteolytic system to adaptation failure in ECs. In this review, we summarize our current knowledge of calpain–calpastatin-mediated physiologic and pathogenic regulation in ECs and discuss the molecular basis by which disruption of this system perturbs EC adaptation to the inflammatory environment.
Collapse
|
29
|
Abstract
Epithelial cells form highly organized polarized sheets with characteristic cell morphologies and tissue architecture. Cell–cell adhesion and intercellular communication are prerequisites of such cohesive sheets of cells, and cell connectivity is mediated through several junctional assemblies, namely desmosomes, adherens, tight and gap junctions. These cell–cell junctions form signalling hubs that not only mediate cell–cell adhesion but impact on multiple aspects of cell behaviour, helping to coordinate epithelial cell shape, polarity and function. This review will focus on the tight and adherens junctions, constituents of the apical junctional complex, and aims to provide a comprehensive overview of the complex signalling that underlies junction assembly, integrity and plasticity.
Collapse
Affiliation(s)
- Alexandra D Rusu
- School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK
| | - Marios Georgiou
- School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK
| |
Collapse
|
30
|
Dókus LE, Yousef M, Bánóczi Z. Modulators of calpain activity: inhibitors and activators as potential drugs. Expert Opin Drug Discov 2020; 15:471-486. [DOI: 10.1080/17460441.2020.1722638] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Levente Endre Dókus
- Department of Organic Chemistry, Institute of Chemistry, Eötvös Loránd University, Budapest, Hungary
| | - Mo’ath Yousef
- Department of Organic Chemistry, Institute of Chemistry, Eötvös Loránd University, Budapest, Hungary
| | - Zoltán Bánóczi
- Department of Organic Chemistry, Institute of Chemistry, Eötvös Loránd University, Budapest, Hungary
| |
Collapse
|
31
|
Rausch V, Hansen CG. The Hippo Pathway, YAP/TAZ, and the Plasma Membrane. Trends Cell Biol 2019; 30:32-48. [PMID: 31806419 DOI: 10.1016/j.tcb.2019.10.005] [Citation(s) in RCA: 176] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 10/13/2019] [Accepted: 10/14/2019] [Indexed: 12/14/2022]
Abstract
The plasma membrane allows the cell to sense and adapt to changes in the extracellular environment by relaying external inputs via intracellular signaling networks. One central cellular signaling pathway is the Hippo pathway, which regulates homeostasis and plays chief roles in carcinogenesis and regenerative processes. Recent studies have found that mechanical stimuli and diffusible chemical components can regulate the Hippo pathway primarily through receptors embedded in the plasma membrane. Morphologically defined structures within the plasma membrane, such as cellular junctions, focal adhesions, primary cilia, caveolae, clathrin-coated pits, and plaques play additional key roles. Here, we discuss recent evidence highlighting the importance of these specialized plasma membrane domains in cellular feedback via the Hippo pathway.
Collapse
Affiliation(s)
- Valentina Rausch
- Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh bioQuarter, 47 Little France Crescent, Edinburgh EH16 4TJ, UK; Institute for Regeneration and Repair, University of Edinburgh, Edinburgh bioQuarter, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Carsten G Hansen
- Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh bioQuarter, 47 Little France Crescent, Edinburgh EH16 4TJ, UK; Institute for Regeneration and Repair, University of Edinburgh, Edinburgh bioQuarter, 5 Little France Drive, Edinburgh EH16 4UU, UK.
| |
Collapse
|
32
|
Pierce RW. Letter in response to “Vascular endothelial cadherin shedding is more severe in sepsis patients with severe acute kidney injury”. Crit Care 2019; 23:167. [PMID: 31088576 PMCID: PMC6515592 DOI: 10.1186/s13054-019-2455-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 04/25/2019] [Indexed: 11/16/2022] Open
|
33
|
Tejeda GS, Esteban‐Ortega GM, San Antonio E, Vidaurre ÓG, Díaz‐Guerra M. Prevention of excitotoxicity-induced processing of BDNF receptor TrkB-FL leads to stroke neuroprotection. EMBO Mol Med 2019; 11:e9950. [PMID: 31273936 PMCID: PMC6609917 DOI: 10.15252/emmm.201809950] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 05/02/2019] [Accepted: 05/03/2019] [Indexed: 12/14/2022] Open
Abstract
Neuroprotective strategies aimed to pharmacologically treat stroke, a prominent cause of death, disability, and dementia, have remained elusive. A promising approach is restriction of excitotoxic neuronal death in the infarct penumbra through enhancement of survival pathways initiated by brain-derived neurotrophic factor (BDNF). However, boosting of neurotrophic signaling after ischemia is challenged by downregulation of BDNF high-affinity receptor, full-length tropomyosin-related kinase B (TrkB-FL), due to calpain-degradation, and, secondarily, regulated intramembrane proteolysis. Here, we have designed a blood-brain barrier (BBB) permeable peptide containing TrkB-FL sequences (TFL457 ) which prevents receptor disappearance from the neuronal surface, early induced after excitotoxicity. In this way, TFL457 interferes TrkB-FL cleavage by both proteolytic systems and increases neuronal viability via a PLCγ-dependent mechanism. By preserving downstream CREB and MEF2 promoter activities, TFL457 initiates a feedback mechanism favoring increased levels in excitotoxic neurons of critical prosurvival mRNAs and proteins. This neuroprotective peptide could be highly relevant for stroke therapy since, in a mouse ischemia model, it counteracts TrkB-FL downregulation in the infarcted brain, efficiently decreases infarct size, and improves neurological outcome.
Collapse
Affiliation(s)
- Gonzalo S Tejeda
- Instituto de Investigaciones Biomédicas “Alberto Sols”Consejo Superior de Investigaciones Científicas‐Universidad Autónoma de Madrid (CSIC‐UAM)MadridSpain
- Present address:
Gardiner LaboratoryInstitute of Cardiovascular and Medical SciencesCollege of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Gema M Esteban‐Ortega
- Instituto de Investigaciones Biomédicas “Alberto Sols”Consejo Superior de Investigaciones Científicas‐Universidad Autónoma de Madrid (CSIC‐UAM)MadridSpain
| | - Esther San Antonio
- Instituto de Investigaciones Biomédicas “Alberto Sols”Consejo Superior de Investigaciones Científicas‐Universidad Autónoma de Madrid (CSIC‐UAM)MadridSpain
| | - Óscar G Vidaurre
- Instituto de Investigaciones Biomédicas “Alberto Sols”Consejo Superior de Investigaciones Científicas‐Universidad Autónoma de Madrid (CSIC‐UAM)MadridSpain
| | - Margarita Díaz‐Guerra
- Instituto de Investigaciones Biomédicas “Alberto Sols”Consejo Superior de Investigaciones Científicas‐Universidad Autónoma de Madrid (CSIC‐UAM)MadridSpain
| |
Collapse
|
34
|
Endothelial cell Piezo1 mediates pressure-induced lung vascular hyperpermeability via disruption of adherens junctions. Proc Natl Acad Sci U S A 2019; 116:12980-12985. [PMID: 31186359 PMCID: PMC6600969 DOI: 10.1073/pnas.1902165116] [Citation(s) in RCA: 165] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Increased hydrostatic pressure in lung capillaries experienced during high altitude, head trauma, and left heart failure can lead to disruption of lung endothelial barrier and edema formation. We identified Piezo1 as a mechanical sensor responsible for endothelial barrier breakdown (barotrauma) secondary to reduced expression of the endothelial adherens junction proteins VE-cadherin, β-catenin, and p120-catenin. Endothelial-specific deletion or pharmacological inhibition of Piezo1 prevented lung capillary leakage, suggesting a therapeutic approach for preventing edema and associated lung failure. Increased pulmonary microvessel pressure experienced in left heart failure, head trauma, or high altitude can lead to endothelial barrier disruption referred to as capillary “stress failure” that causes leakage of protein-rich plasma and pulmonary edema. However, little is known about vascular endothelial sensing and transduction of mechanical stimuli inducing endothelial barrier disruption. Piezo1, a mechanosensing ion channel expressed in endothelial cells (ECs), is activated by elevated pressure and other mechanical stimuli. Here, we demonstrate the involvement of Piezo1 in sensing increased lung microvessel pressure and mediating endothelial barrier disruption. Studies were made in mice in which Piezo1 was deleted conditionally in ECs (Piezo1iΔEC), and lung microvessel pressure was increased either by raising left atrial pressure or by aortic constriction. We observed that lung endothelial barrier leakiness and edema induced by raising pulmonary microvessel pressure were abrogated in Piezo1iΔEC mice. Piezo1 signaled lung vascular hyperpermeability by promoting the internalization and degradation of the endothelial adherens junction (AJ) protein VE-cadherin. Breakdown of AJs was the result of activation of the calcium-dependent protease calpain and degradation of the AJ proteins VE-cadherin, β-catenin, and p120-catenin. Deletion of Piezo1 in ECs or inhibition of calpain similarly prevented reduction in the AJ proteins. Thus, Piezo1 activation in ECs induced by elevated lung microvessel pressure mediates capillary stress failure and edema formation secondary to calpain-induced disruption of VE-cadherin adhesion. Inhibiting Piezo1 signaling may be a useful strategy to limit lung capillary stress failure injury in response to elevated vascular pressures.
Collapse
|
35
|
Wu MA, Tsvirkun D, Bureau L, Boccon-Gibod I, Inglebert M, Duperray A, Bouillet L, Misbah C, Cicardi M. Paroxysmal Permeability Disorders: Development of a Microfluidic Device to Assess Endothelial Barrier Function. Front Med (Lausanne) 2019; 6:89. [PMID: 31069229 PMCID: PMC6491734 DOI: 10.3389/fmed.2019.00089] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 04/09/2019] [Indexed: 11/28/2022] Open
Abstract
Background: Paroxysmal Permeability Disorders (PPDs) are pathological conditions caused by periodic short lasting increase of endothelial permeability, in the absence of inflammatory, degenerative, ischemic vascular injury. PPDs include primary angioedema, idiopathic systemic capillary leak syndrome and some rare forms of localized retroperitoneal-mediastinal edema. Aim: to validate a microfluidic device to study endothelial permeability in flow conditions. Materials and Methods: we designed a microchannel network (the smallest channel is 30μm square section). Human Umbilical Vein Endothelial Cells (HUVECs) were cultured under constant shear stress in the networks. Endothelial permeability assessment was based on interaction of biotinylated fibronectin used as a matrix for HUVECs and FITC-conjugated avidin. The increase in endothelial permeability was identified as changes in fluorescence intensity detected by confocal fluorescent microscopy. Results: The microchannels were constantly perfused with a steady flow of culture medium, ensuring a physiologically relevant level of shear stress at the wall of ~0.2 Pa. Our preliminary results demonstrated that circulation of culture medium or plasma from healthy volunteers was associated with low fluorescence of fibronectin matrix. When bradykinin diluted in culture medium was perfused, an increase in average fluorescence was detected. Conclusion: Our microvasculature model is suitable to study endothelial functions in physiological flow conditions and in the presence of factors like bradykinin known as mediator of several PPDs. Therefore, it can be a promising tool to better understand the mechanisms underlying disorders of endothelial permeability.
Collapse
Affiliation(s)
- Maddalena Alessandra Wu
- Department of Biomedical and Clinical Sciences, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, University of Milan, Milan, Italy
| | - Daria Tsvirkun
- Univ. Grenoble Alpes, LIPHY, Grenoble, France.,CNRS, LIPHY, Grenoble, France.,Belozersky Institute of Physico-chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Lionel Bureau
- Univ. Grenoble Alpes, LIPHY, Grenoble, France.,CNRS, LIPHY, Grenoble, France
| | - Isabelle Boccon-Gibod
- Department of Internal Medicine, Centre Hospitalier Universitaire de Grenoble, Grenoble, France
| | | | - Alain Duperray
- Univ. Grenoble Alpes, IAB, Grenoble, France.,INSERM, IAB, Grenoble, France
| | - Laurence Bouillet
- Department of Internal Medicine, Centre Hospitalier Universitaire de Grenoble, Grenoble, France
| | - Chaouqi Misbah
- Univ. Grenoble Alpes, LIPHY, Grenoble, France.,CNRS, LIPHY, Grenoble, France
| | - Marco Cicardi
- IRCCS-Istituti Clinici Scientifici Maugeri, University of Milan, Milan, Italy
| |
Collapse
|
36
|
Liao D, Mei H, Hu Y, Newman DK, Newman PJ. CRISPR-mediated deletion of the PECAM-1 cytoplasmic domain increases receptor lateral mobility and strengthens endothelial cell junctional integrity. Life Sci 2018; 193:186-193. [PMID: 29122551 DOI: 10.1016/j.lfs.2017.11.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 10/24/2017] [Accepted: 11/04/2017] [Indexed: 10/18/2022]
Abstract
AIMS PECAM-1 is an abundant endothelial cell surface receptor that becomes highly enriched at endothelial cell-cell junctions, where it functions to mediate leukocyte transendothelial migration, sense changes in shear and flow, and maintain the vascular permeability barrier. Homophilic interactions mediated by the PECAM-1 extracellular domain are known to be required for PECAM-1 to perform these functions; however, much less is understood about the role of its cytoplasmic domain in these processes. MAIN METHODS CRISPR/Cas9 gene editing technology was employed to generate human endothelial cell lines that either lack PECAM-1 entirely, or express mutated PECAM-1 missing the majority of its cytoplasmic domain (∆CD-PECAM-1). The endothelial barrier function was evaluated by Electric Cell-substrate Impedance Sensing, and molecular mobility was assessed by fluorescence recovery after photobleaching. KEY FINDINGS We found that ∆CD-PECAM-1 concentrates normally at endothelial cell junctions, but has the unexpected property of conferring increased baseline barrier resistance, as well as a more rapid rate of recovery of vascular integrity following thrombin-induced disruption of the endothelial barrier. Fluorescence recovery after photobleaching analysis revealed that ∆CD-PECAM-1 exhibits increased mobility within the plane of the plasma membrane, thus allowing it to redistribute more rapidly back to endothelial cell-cell borders to reform the vascular permeability barrier. SIGNIFICANCE The PECAM-1 cytoplasmic domain plays a novel role in regulating the rate and extent of vascular permeability following thrombotic or inflammatory challenge.
Collapse
Affiliation(s)
- Danying Liao
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI, United States; Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Heng Mei
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Debra K Newman
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI, United States; Department of Pharmacology, Medical College of Wisconsin, Milwaukee, United States; Department of Microbiology, Medical College of Wisconsin, Milwaukee, United States; The Cardiovascular Center, Medical College of Wisconsin, Milwaukee, United States
| | - Peter J Newman
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI, United States; Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Department of Pharmacology, Medical College of Wisconsin, Milwaukee, United States; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, United States; The Cardiovascular Center, Medical College of Wisconsin, Milwaukee, United States.
| |
Collapse
|
37
|
Abstract
Under physiological conditions, the arterial endothelium exerts a powerful protective influence to maintain vascular homeostasis. However, during the development of vascular disease, these protective activities are lost, and dysfunctional endothelial cells actually promote disease pathogenesis. Numerous investigations have analyzed the characteristics of dysfunctional endothelium with a view to understanding the processes responsible for the dysfunction and to determining their role in vascular pathology. This review adopts an alternate approach: reviewing the mechanisms that contribute to the initial formation of a healthy protective endothelium and on how those mechanisms may be disrupted, precipitating the appearance of dysfunctional endothelial cells and the progression of vascular disease. This approach, which highlights the role of endothelial adherens junctions and vascular endothelial-cadherin in endothelial maturation and endothelial dysfunction, provides new insight into the remarkable biology of this important cell layer and its role in vascular protection and vascular disease.
Collapse
|
38
|
Lei X, Chen M, Huang M, Li X, Shi C, Zhang D, Luo L, Zhang Y, Ma N, Chen H, Liang H, Ye W, Zhang D. Desacetylvinblastine Monohydrazide Disrupts Tumor Vessels by Promoting VE-cadherin Internalization. Am J Cancer Res 2018; 8:384-398. [PMID: 29290815 PMCID: PMC5743555 DOI: 10.7150/thno.22222] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 10/13/2017] [Indexed: 01/18/2023] Open
Abstract
Vinca alkaloids, the well-known tubulin-binding agents, are widely used for the clinical treatment of malignant tumors. However, little attention has been paid to their vascular disrupting effects, and the underlying mechanisms remain largely unknown. This study aims to investigate the vascular disrupting effect and the underlying mechanisms of vinca alkaloids. Methods: The capillary disruption assay and aortic ring assay were performed to evaluate the in vitro vascular disrupting effect of desacetylvinblastine monohydrazide (DAVLBH), a derivate of vinblastine, and the in vivo vascular disrupting effect was assessed on HepG2 xenograft model using magnetic resonance imaging, hematoxylin and eosin staining and immunohistochemistry. Tubulin polymerization, endothelial cell monolayer permeability, western blotting and immunofluorescence assays were performed to explore the underlying mechanisms of DAVLBH-mediated tumor vascular disruption. Results: DAVLBH has potent vascular disrupting activity both in vitro and in vivo. DAVLBH disrupts tumor vessels in a different manner than classical tubulin-targeting VDAs; it inhibits microtubule polymerization, promotes the internalization of vascular endothelial cadherin (VE-cadherin) and inhibits the recycling of internalized VE-cadherin to the cell membrane, thus increasing endothelial cell permeability and ultimately resulting in vascular disruption. DAVLBH-mediated promotion of VE-cadherin internalization and inhibition of internalized VE-cadherin recycling back to the cell membrane are partly dependent on inhibition of microtubule polymerization, and Src activation is involved in DAVLBH-induced VE-cadherin internalization. Conclusions: This study sheds light on the tumor vascular disrupting effect and underlying mechanisms of vinca alkaloids and provides new insight into the molecular mechanism of tubulin-targeting VDAs.
Collapse
|
39
|
Lapaquette P, Fritah S, Lhocine N, Andrieux A, Nigro G, Mounier J, Sansonetti P, Dejean A. Shigella entry unveils a calcium/calpain-dependent mechanism for inhibiting sumoylation. eLife 2017; 6:27444. [PMID: 29231810 PMCID: PMC5745084 DOI: 10.7554/elife.27444] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 12/11/2017] [Indexed: 12/28/2022] Open
Abstract
Disruption of the sumoylation/desumoylation equilibrium is associated with several disease states such as cancer and infections, however the mechanisms regulating the global SUMO balance remain poorly defined. Here, we show that infection by Shigella flexneri, the causative agent of human bacillary dysentery, switches off host sumoylation during epithelial cell infection in vitro and in vivo and that this effect is mainly mediated by a calcium/calpain-induced cleavage of the SUMO E1 enzyme SAE2, thus leading to sumoylation inhibition. Furthermore, we describe a mechanism by which Shigella promotes its own invasion by altering the sumoylation state of RhoGDIα, a master negative regulator of RhoGTPase activity and actin polymerization. Together, our data suggest that SUMO modification is essential to restrain pathogenic bacterial entry by limiting cytoskeletal rearrangement induced by bacterial effectors. Moreover, these findings identify calcium-activated calpains as powerful modulators of cellular sumoylation levels with potentially broad implications in several physiological and pathological situations.
Collapse
Affiliation(s)
- Pierre Lapaquette
- Nuclear Organization and Oncogenesis Unit, Institut Pasteur, Paris, France.,INSERM, U993, Paris, France
| | - Sabrina Fritah
- Nuclear Organization and Oncogenesis Unit, Institut Pasteur, Paris, France.,INSERM, U993, Paris, France
| | - Nouara Lhocine
- Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, Paris, France.,INSERM, U786, Paris, France
| | - Alexandra Andrieux
- Nuclear Organization and Oncogenesis Unit, Institut Pasteur, Paris, France.,INSERM, U993, Paris, France
| | - Giulia Nigro
- Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, Paris, France.,INSERM, U786, Paris, France
| | - Joëlle Mounier
- Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, Paris, France.,INSERM, U786, Paris, France
| | - Philippe Sansonetti
- Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, Paris, France.,INSERM, U786, Paris, France
| | - Anne Dejean
- Nuclear Organization and Oncogenesis Unit, Institut Pasteur, Paris, France.,INSERM, U993, Paris, France
| |
Collapse
|
40
|
Tompkins N, Spinelli KJ, Choi D, Barr-Gillespie PG. A Model for Link Pruning to Establish Correctly Polarized and Oriented Tip Links in Hair Bundles. Biophys J 2017; 113:1868-1881. [PMID: 29045880 DOI: 10.1016/j.bpj.2017.08.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 08/14/2017] [Accepted: 08/21/2017] [Indexed: 10/18/2022] Open
Abstract
Tip links are thought to gate the mechanically sensitive transduction channels of hair cells, but how they form during development and regeneration remains mysterious. In particular, it is unclear how tip links are strung between stereocilia so that they are oriented parallel to a single axis; why their polarity is uniform despite their constituent molecules' intrinsic asymmetry; and why only a single tip link is present at each tip-link position. We present here a series of simple rules that reasonably explain why these phenomena occur. In particular, our model relies on each of the two ends of the tip link having distinct Ca2+-dependent stability and being connected to different motor complexes. A simulation employing these rules allowed us to explore the parameter space for the model, demonstrating the importance of the feedback between transduction channels and angled links, links that are 60° off-axis with respect to mature tip links. We tested this key aspect of the model by examining angled links in chick cochlea hair cells. As implied by the assumptions used to generate the model, we found that angled links were stabilized if there was no tip link at the tip of the upper stereocilium, and appeared when transduction channels were blocked. The model thus plausibly explains how tip-link formation and pruning can occur.
Collapse
Affiliation(s)
- Nathan Tompkins
- Oregon Hearing Research Center and Vollum Institute, Oregon Health and Science University, Portland, Oregon
| | - Kateri J Spinelli
- Oregon Hearing Research Center and Vollum Institute, Oregon Health and Science University, Portland, Oregon
| | - Dongseok Choi
- School of Public Health, Oregon Health and Science University, Portland, Oregon; Graduate School of Dentistry, Kyung Hee University, Seoul, South Korea
| | - Peter G Barr-Gillespie
- Oregon Hearing Research Center and Vollum Institute, Oregon Health and Science University, Portland, Oregon.
| |
Collapse
|
41
|
Mendoza PA, Silva P, Díaz J, Arriagada C, Canales J, Cerda O, Torres VA. Calpain2 mediates Rab5-driven focal adhesion disassembly and cell migration. Cell Adh Migr 2017; 12:185-194. [PMID: 29099266 DOI: 10.1080/19336918.2017.1377388] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The early endosome protein Rab5 was recently shown to promote cell migration by enhancing focal adhesion disassembly through mechanisms that remain elusive. Focal adhesion disassembly is associated to proteolysis of talin, in a process that requires calpain2. Since calpain2 has been found at vesicles and endosomal compartments, we hypothesized that Rab5 stimulates calpain2 activity, leading to enhanced focal adhesion disassembly in migrating cells. We observed that calpain2 co-localizes with EEA1-positive early endosomes and co-immunoprecipitates with EEA1 and Rab5 in A549 lung carcinoma cells undergoing spreading, whereas Rab5 knock-down decreased the accumulation of calpain2 at early endosomal-enriched fractions. In addition, Rab5 silencing decreased calpain2 activity, as shown by cleavage of the fluorogenic substrate tBOC-LM-CMAC and the endogenous substrate talin. Accordingly, Rab5 promoted focal adhesion disassembly in a calpain2-dependent manner, as expression of GFP-Rab5 accelerated focal adhesion disassembly in nocodazole-synchronized cells, whereas pharmacological inhibition of calpain2 with N-acetyl-Leu-Leu-Met prevented both focal adhesion disassembly and cell migration induced by Rab5. In summary, these data uncover Rab5 as a novel regulator of calpain2 activity and focal adhesion proteolysis leading to cell migration.
Collapse
Affiliation(s)
- Pablo A Mendoza
- a Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile , Santiago , Chile.,b Molecular Pathology Laboratory , Institute of Biochemistry and Microbiology, Sciences Faculty, Universidad Austral de Chile , Valdivia , Chile
| | - Patricio Silva
- a Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile , Santiago , Chile.,c Faculty of Health Sciences, Universidad Central de Chile , Santiago , Chile
| | - Jorge Díaz
- a Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile , Santiago , Chile.,d Advanced Center for Chronic Diseases (ACCDiS) , Universidad de Chile , Santiago , Chile
| | - Cecilia Arriagada
- a Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile , Santiago , Chile
| | - Jimena Canales
- e Programa de Biología Celular y Molecular , Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile , Santiago , Chile
| | - Oscar Cerda
- e Programa de Biología Celular y Molecular , Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile , Santiago , Chile.,f Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD) , Universidad de Chile , Santiago , Chile
| | - Vicente A Torres
- a Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile , Santiago , Chile.,d Advanced Center for Chronic Diseases (ACCDiS) , Universidad de Chile , Santiago , Chile
| |
Collapse
|
42
|
IP 3 receptor signaling and endothelial barrier function. Cell Mol Life Sci 2017; 74:4189-4207. [PMID: 28803370 DOI: 10.1007/s00018-017-2624-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 07/18/2017] [Accepted: 08/08/2017] [Indexed: 12/14/2022]
Abstract
The endothelium, a monolayer of endothelial cells lining vessel walls, maintains tissue-fluid homeostasis by restricting the passage of the plasma proteins and blood cells into the interstitium. The ion Ca2+, a ubiquitous secondary messenger, initiates signal transduction events in endothelial cells that is critical to control of vascular tone and endothelial permeability. The ion Ca2+ is stored inside the intracellular organelles and released into the cytosol in response to environmental cues. The inositol 1,4,5-trisphosphate (IP3) messenger facilitates Ca2+ release through IP3 receptors which are Ca2+-selective intracellular channels located within the membrane of the endoplasmic reticulum. Binding of IP3 to the IP3Rs initiates assembly of IP3R clusters, a key event responsible for amplification of Ca2+ signals in endothelial cells. This review discusses emerging concepts related to architecture and dynamics of IP3R clusters, and their specific role in propagation of Ca2+ signals in endothelial cells.
Collapse
|
43
|
Urbano RL, Furia C, Basehore S, Clyne AM. Stiff Substrates Increase Inflammation-Induced Endothelial Monolayer Tension and Permeability. Biophys J 2017; 113:645-655. [PMID: 28793219 PMCID: PMC5550298 DOI: 10.1016/j.bpj.2017.06.033] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 05/15/2017] [Accepted: 06/13/2017] [Indexed: 01/22/2023] Open
Abstract
Arterial stiffness and inflammation are associated with atherosclerosis, and each have individually been shown to increase endothelial monolayer tension and permeability. The objective of this study was to determine if substrate stiffness enhanced endothelial monolayer tension and permeability in response to inflammatory cytokines. Porcine aortic endothelial cells were cultured at confluence on polyacrylamide gels of varying stiffness and treated with either tumor necrosis factor-α (TNFα) or thrombin. Monolayer tension was measured through vinculin localization at the cell membrane, traction force microscopy, and phosphorylated myosin light chain quantity and actin fiber colocalization. Cell permeability was measured by cell-cell junction confocal microscopy and a dextran permeability assay. When treated with TNFα or thrombin, endothelial monolayers on stiffer substrates showed increased traction forces, vinculin at the cell membrane, and vinculin phosphorylation, suggesting elevated monolayer tension. Interestingly, VE-cadherin shifted toward a smaller molecular weight in endothelial monolayers on softer substrates, which may relate to increased VE-cadherin endocytosis and degradation. Phosphorylated myosin light chain colocalization with actin stress fibers increased in endothelial monolayers treated with TNFα or thrombin on stiffer substrates, indicating elevated cell monolayer contractility. Endothelial monolayers also developed focal adherens intercellular junctions and became more permeable when cultured on stiffer substrates in the presence of the inflammatory cytokines. Whereas each of these effects was likely mitigated by Rho/ROCK, Rho/ROCK pathway inhibition via Y27632 disrupted cell-cell junction morphology, showing that cell contractility is required to maintain adherens junction integrity. These data suggest that stiff substrates change intercellular junction protein localization and degradation, which may counteract the inflammation-induced increase in endothelial monolayer tension and thereby moderate inflammation-induced junction loss and associated endothelial monolayer permeability on stiffer substrates.
Collapse
|
44
|
Chang F, Flavahan S, Flavahan NA. Impaired activity of adherens junctions contributes to endothelial dilator dysfunction in ageing rat arteries. J Physiol 2017; 595:5143-5158. [PMID: 28561330 DOI: 10.1113/jp274189] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 05/17/2017] [Indexed: 12/18/2022] Open
Abstract
KEY POINTS Ageing-induced endothelial dysfunction contributes to organ dysfunction and progression of cardiovascular disease. VE-cadherin clustering at adherens junctions promotes protective endothelial functions, including endothelium-dependent dilatation. Ageing increased internalization and degradation of VE-cadherin, resulting in impaired activity of adherens junctions. Inhibition of VE-cadherin clustering at adherens junctions (function-blocking antibody; FBA) reduced endothelial dilatation in young arteries but did not affect the already impaired dilatation in old arteries. After junctional disruption with the FBA, dilatation was similar in young and old arteries. Src tyrosine kinase activity and tyrosine phosphorylation of VE-cadherin were increased in old arteries. Src inhibition increased VE-cadherin at adherens junctions and increased endothelial dilatation in old, but not young, arteries. Src inhibition did not increase dilatation in old arteries treated with the VE-cadherin FBA. Ageing impairs the activity of adherens junctions, which contributes to endothelial dilator dysfunction. Restoring the activity of adherens junctions could be of therapeutic benefit in vascular ageing. ABSTRACT Endothelial dilator dysfunction contributes to pathological vascular ageing. Experiments assessed whether altered activity of endothelial adherens junctions (AJs) might contribute to this dysfunction. Aortas and tail arteries were isolated from young (3-4 months) and old (22-24 months) F344 rats. VE-cadherin immunofluorescent staining at endothelial AJs and AJ width were reduced in old compared to young arteries. A 140 kDa VE-cadherin species was present on the cell surface and in TTX-insoluble fractions, consistent with junctional localization. Levels of the 140 kDa VE-cadherin were decreased, whereas levels of a TTX-soluble 115 kDa VE-cadherin species were increased in old compared to young arteries. Acetylcholine caused endothelium-dependent dilatation that was decreased in old compared to young arteries. Disruption of VE-cadherin clustering at AJs (function-blocking antibody, FBA) inhibited dilatation to acetylcholine in young, but not old, arteries. After the FBA, there was no longer any difference in dilatation between old and young arteries. Src activity and tyrosine phosphorylation of VE-cadherin were increased in old compared to young arteries. In old arteries, Src inhibition (saracatinib) increased: (i) 140 kDa VE-cadherin in the TTX-insoluble fraction, (ii) VE-cadherin intensity at AJs, (iii) AJ width, and (iv) acetylcholine dilatation. In old arteries treated with the FBA, saracatinib no longer increased acetylcholine dilatation. Saracatinib did not affect dilatation in young arteries. Therefore, ageing impairs AJ activity, which appears to reflect Src-induced phosphorylation, internalization and degradation of VE-cadherin. Moreover, impaired AJ activity can account for the endothelial dilator dysfunction in old arteries. Restoring endothelial AJ activity may be a novel therapeutic approach to vascular ageing.
Collapse
Affiliation(s)
- Fumin Chang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Sheila Flavahan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Nicholas A Flavahan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|