1
|
Kizhatil K, Clark GM, Sunderland DK, Bhandari A, Horbal LJ, Balasubramanian R, John SWM. FYN regulates aqueous humor outflow and IOP through the phosphorylation of VE-CADHERIN. Nat Commun 2025; 16:51. [PMID: 39746990 PMCID: PMC11696269 DOI: 10.1038/s41467-024-55232-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 12/03/2024] [Indexed: 01/04/2025] Open
Abstract
Schlemm's canal endothelial cells (SECs) serve as the final barrier to aqueous humor (AQH) drainage from the eye. SECs adjust permeability to AQH outflow to modulate intraocular pressure (IOP). The broad identification of IOP-related genes implicates SECs in glaucoma. However, the molecular mechanisms by which SECs sense and respond to pressure changes to regulate fluid permeability and IOP remain largely undefined. We hypothesize that mechano-responsive phosphorylation of the cell adhesion molecule VE-CADHERIN (CDH5) in SECs, by FYN and possibly other SRC family kinases, regulates adherens junction (AJ) permeability to AQH in response to IOP. On experimentally raising IOP in mouse eyes, AJ permeability, CDH5 phosphorylation, and FYN activation at the AJ all increase. FYN null mutant mice display disrupted IOP regulation and reduced AQH outflow. These findings demonstrate an important role of mechanotransducive signaling within SECs in maintaining IOP homeostasis and implicate FYN as a potential target for developing IOP-lowering treatments.
Collapse
Affiliation(s)
- Krishnakumar Kizhatil
- Department of Ophthalmology and Visual Sciences, The Ohio State Medical Center, Columbus, Ohio, 43210, USA.
| | | | | | - Aakriti Bhandari
- The Jackson Laboratory, Bar Harbor, Maine, 04609, USA
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY, USA
| | - Logan J Horbal
- The Jackson Laboratory, Bar Harbor, Maine, 04609, USA
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Simon W M John
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY, USA.
- Zuckerman Mind Brain Behavior Institute, New York, NY, 10027, USA.
| |
Collapse
|
2
|
Wang L, Chen Y, Wu H, Yu HH, Ma L. Slit2-Robo4 signal pathway and tight junction in intestine mediate LPS-induced inflammation in mice. Eur J Med Res 2024; 29:349. [PMID: 38937814 PMCID: PMC11209965 DOI: 10.1186/s40001-024-01894-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 05/21/2024] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND Sepsis is one of the most common clinical diseases, which is characterized by a serious and uncontrollable inflammatory response. LPS-induced inflammation is a critical pathological event in sepsis, but the underlying mechanism has not yet been fully elucidated. METHODS The animal model was established for two batches. In the first batch of experiments, Adult C57BL/6J mice were randomly divided into control group and LPS (5 mg/kg, i.p.)group . In the second batch of experiments, mice were randomly divided into control group, LPS group, and LPS+VX765(10 mg/kg, i.p., an inhibitor of NLRP3 inflammasome) group. After 24 hours, mice were anesthetized with isoflurane, blood and intestinal tissue were collected for tissue immunohistochemistry, Western blot analysis and ELISA assays. RESULTS The C57BL/6J mice injected with LPS for twenty-four hours could exhibit severe inflammatory reaction including an increased IL-1β, IL-18 in serum and activation of NLRP3 inflammasome in intestine. The injection of VX765 could reverse these effects induced by LPS. These results indicated that the increased level of IL-1β and IL-18 in serum induced by LPS is related to the increased intestinal permeability and activation of NLRP3 inflammasome. In the second batch of experiments, results of western blot and immunohistochemistry showed that Slit2 and Robo4 were significant decreased in intestine of LPS group, while the expression of VEGF was significant increased. Meanwhile, the protein level of tight junction protein ZO-1, occludin, and claudin-5 were significantly lower than in control group, which could also be reversed by VX765 injection. CONCLUSIONS In this study, we revealed that Slit2-Robo4 signaling pathway and tight junction in intestine may be involved in LPS-induced inflammation in mice, which may account for the molecular mechanism of sepsis.
Collapse
Affiliation(s)
- Lv Wang
- Department of Emergency and Critical Care Medicine, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, People's Republic of China
| | - Yingtai Chen
- Emergency Department, Baoshan Branch of Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200444, People's Republic of China
| | - Hao Wu
- Department of Emergency and Critical Care Medicine, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, People's Republic of China
| | - He-Hua Yu
- Department of Emergency and Critical Care Medicine, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, People's Republic of China.
| | - Linhao Ma
- Department of Emergency and Critical Care Medicine, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, People's Republic of China.
| |
Collapse
|
3
|
Kryvenko V, Vadász I. Alveolar-capillary endocytosis and trafficking in acute lung injury and acute respiratory distress syndrome. Front Immunol 2024; 15:1360370. [PMID: 38533500 PMCID: PMC10963603 DOI: 10.3389/fimmu.2024.1360370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 02/29/2024] [Indexed: 03/28/2024] Open
Abstract
Acute respiratory distress syndrome (ARDS) is associated with high morbidity and mortality but lacks specific therapeutic options. Diverse endocytic processes play a key role in all phases of acute lung injury (ALI), including the initial insult, development of respiratory failure due to alveolar flooding, as a consequence of altered alveolar-capillary barrier function, as well as in the resolution or deleterious remodeling after injury. In particular, clathrin-, caveolae-, endophilin- and glycosylphosphatidyl inositol-anchored protein-mediated endocytosis, as well as, macropinocytosis and phagocytosis have been implicated in the setting of acute lung damage. This manuscript reviews our current understanding of these endocytic pathways and subsequent intracellular trafficking in various phases of ALI, and also aims to identify potential therapeutic targets for patients with ARDS.
Collapse
Affiliation(s)
- Vitalii Kryvenko
- Department of Internal Medicine, Justus Liebig University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
- The Cardio-Pulmonary Institute (CPI), Giessen, Germany
- Institute for Lung Health (ILH), Giessen, Germany
| | - István Vadász
- Department of Internal Medicine, Justus Liebig University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
- The Cardio-Pulmonary Institute (CPI), Giessen, Germany
- Institute for Lung Health (ILH), Giessen, Germany
| |
Collapse
|
4
|
Kizhatil K, Clark G, Sunderland D, Bhandari A, Horbal L, Balasubramanian R, John S. FYN regulates aqueous humor outflow and IOP through the phosphorylation of VE-cadherin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.04.556253. [PMID: 37886565 PMCID: PMC10602025 DOI: 10.1101/2023.09.04.556253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
The exact sites and molecules that determine resistance to aqueous humor drainage and control intraocular pressure (IOP) need further elaboration. Proposed sites include the inner wall of Schlemms's canal and the juxtacanalicular trabecular meshwork ocular drainage tissues. The adherens junctions (AJs) of Schlemm's canal endothelial cells (SECs) must both preserve the blood-aqueous humor (AQH) barrier and be conducive to AQH drainage. How homeostatic control of AJ permeability in SC occurs and how such control impacts IOP is unclear. We hypothesized that mechano-responsive phosphorylation of the junctional molecule VE-CADHERIN (VEC) by SRC family kinases (SFKs) regulates the permeability of SEC AJs. We tested this by clamping IOP at either 16 mmHg, 25 mmHg, or 45 mmHg in mice and then measuring AJ permeability and VEC phosphorylation. We found that with increasing IOP: 1) SEC AJ permeability increased, 2) VEC phosphorylation was increased at tyrosine-658, and 3) SFKs were activated at the AJ. Among the two SFKs known to phosphorylate VEC, FYN, but not SRC, localizes to the SC. Furthermore, FYN mutant mice had decreased phosphorylation of VEC at SEC AJs, dysregulated IOP, and reduced AQH outflow. Together, our data demonstrate that increased IOP activates FYN in the inner wall of SC, leading to increased phosphorylation of AJ VEC and, thus, decreased resistance to AQH outflow. These findings support a crucial role of mechanotransduction signaling in IOP homeostasis within SC in response to IOP. These data strongly suggest that the inner wall of SC partially contributes to outflow resistance.
Collapse
|
5
|
Locatelli L, Fedele G, Maier JA. The Role of Txnip in Mediating Low-Magnesium-Driven Endothelial Dysfunction. Int J Mol Sci 2023; 24:ijms24098351. [PMID: 37176057 PMCID: PMC10179684 DOI: 10.3390/ijms24098351] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/28/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
Magnesium deficiency is associated with a greater risk of developing cardiovascular diseases since this cation is fundamental in regulating vascular function. This clinical evidence is sustained by in vitro studies showing that culturing endothelial cells in low concentrations of magnesium promotes the acquisition of a pro-oxidant and pro-inflammatory phenotype. Here, we show that the increase in reactive oxygen species in endothelial cells in low-magnesium-containing medium is due to the upregulation of the pro-oxidant protein thioredoxin interacting protein (TXNIP), with a consequent accumulation of lipid droplets and increase in endothelial permeability through the downregulation and relocalization of junctional proteins. Silencing TXNIP restores the endothelial barrier and lipid content. Because (i) mitochondria serve multiple roles in shaping cell function, health and survival and (ii) mitochondria are the main intracellular stores of magnesium, it is of note that no significant alterations were detected in their morphology and dynamics in our experimental model. We conclude that TXNIP upregulation contributes to low-magnesium-induced endothelial dysfunction in vitro.
Collapse
Affiliation(s)
- Laura Locatelli
- Department of Biomedical and Clinical Sciences, Università di Milano, Via GB Grassi 74, 20157 Milano, Italy
| | - Giorgia Fedele
- Department of Biomedical and Clinical Sciences, Università di Milano, Via GB Grassi 74, 20157 Milano, Italy
| | - Jeanette A Maier
- Department of Biomedical and Clinical Sciences, Università di Milano, Via GB Grassi 74, 20157 Milano, Italy
| |
Collapse
|
6
|
Zhao Q, Yao J, Meng B, Shen S, Cao S. Role of the blood-spinal cord barrier: An adheren junction regulation mechanism that promotes chronic postsurgical pain. Biochem Biophys Res Commun 2023; 660:65-72. [PMID: 37068390 DOI: 10.1016/j.bbrc.2023.04.021] [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: 03/02/2023] [Revised: 03/16/2023] [Accepted: 04/10/2023] [Indexed: 04/19/2023]
Abstract
Chronic postsurgical pain (CPSP) is a serious postoperative complication with high incidence, and its pathogenesis involves neuroimmune interactions and the breakdown of the blood-spinal cord barrier (BSCB), the decreased level of adheren junction (AJ)-related proteins is an important cause of BSCB injury. Vascular endothelial-cadherin (VE-cadherin) and p120 catenin (p120) constitute the endothelial barrier adheren junction. The Src/p120/VE-cadherin pathway is involved in the regulation of the endothelial barrier function. However, the role of the BSCB-AJ regulatory mechanism in CPSP has not been reported. In this study, we established a skin/muscle incision and retraction (SMIR) model and evaluated the paw withdrawal threshold (PWT), the effects of an Src inhibitor and p120 knockdown on p-Src, p120 and VE-cadherin expression, as well as BSCB-AJ function in rat spinal cord were observed to explore the regulation of BSCB-AJ function by the p-Src/p120/VE-cadherin pathway in promoting SMIR-induced CPSP. The levels of p-Src, p120 and VE-cadherin in the spinal cord were detected by Western blot. Meanwhile, BSCB permeability test was used to detect the changes in BCSB function. Finally, the spatial and temporal localization of p120 in spinal cord was detected by immunofluorescence. Our findings indicated that p-Src/p120/VE-cadherin could induce BSCB-AJ dysfunction and promote the development of CPSP.
Collapse
Affiliation(s)
- Qihong Zhao
- Medical College, Nantong University, Nantong, Jiangsu, 226001, China
| | - Ju Yao
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong University, Nantong, Jiangsu, 226001, China
| | - Bei Meng
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong University, Nantong, Jiangsu, 226001, China
| | - Shiren Shen
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong University, Nantong, Jiangsu, 226001, China
| | - Su Cao
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong University, Nantong, Jiangsu, 226001, China.
| |
Collapse
|
7
|
Rincon-Benavides MA, Mendonca NC, Cuellar-Gaviria TZ, Salazar-Puerta AI, Ortega-Pineda L, Blackstone BN, Deng B, McComb DW, Gallego-Perez D, Powell HM, Higuita-Castro N. Engineered Vasculogenic Extracellular Vesicles Drive Nonviral Direct Conversions of Human Dermal Fibroblasts into Induced Endothelial Cells and Improve Wound Closure. ADVANCED THERAPEUTICS 2023; 6:2200197. [PMID: 37577183 PMCID: PMC10416766 DOI: 10.1002/adtp.202200197] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Indexed: 08/15/2023]
Abstract
Vasculogenic cell therapies have emerged as a powerful tool to increase vascularization and promote tissue repair/regeneration. Current approaches to cell therapies, however, rely mostly on progenitor cells, which pose significant risks (e.g., uncontrolled differentiation, tumorigenesis, and genetic/epigenetic abnormalities). Moreover, reprogramming methodologies used to generate induced endothelial cells (iECs) from induced pluripotent stem cells rely heavily on viral vectors, which pose additional translational limitations. This work describes the development of engineered human extracellular vesicles (EVs) capable of driving reprogramming-based vasculogenic therapies without the need for progenitor cells and/or viral vectors. The EVs were derived from primary human dermal fibroblasts (HDFs), and were engineered to pack transcription factor genes/transcripts of ETV2, FLI1, and FOXC2 (EFF). Our results indicate that in addition of EFF, the engineered EVs were also loaded with transcripts of angiogenic factors (e.g., VEGF-A, VEGF-KDR, FGF2). In vitro and in vivo studies indicate that such EVs effectively transfected HDFs and drove direct conversions towards iECs within 7-14 days. Finally, wound healing studies in mice indicate that engineered EVs lead to improved wound closure and vascularity. Altogether, our results show the potential of engineered human vasculogenic EVs to drive direct reprogramming processes of somatic cells towards iECs, and facilitate tissue repair/regeneration.
Collapse
Affiliation(s)
- Maria A. Rincon-Benavides
- Biophysics Graduate Program, The Ohio State University, Columbus, OH
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH
| | | | | | | | | | - Britani N. Blackstone
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH
| | - Binbin Deng
- Center for Electron Microscopy and Analysis (CEMAS), The Ohio State University, Columbus, OH
| | - David W McComb
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH
- Center for Electron Microscopy and Analysis (CEMAS), The Ohio State University, Columbus, OH
| | - Daniel Gallego-Perez
- Biophysics Graduate Program, The Ohio State University, Columbus, OH
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH
- Department of Surgery, The Ohio State University, Columbus, OH
| | - Heather M. Powell
- Biophysics Graduate Program, The Ohio State University, Columbus, OH
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH
| | - Natalia Higuita-Castro
- Biophysics Graduate Program, The Ohio State University, Columbus, OH
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH
- Department of Surgery, The Ohio State University, Columbus, OH
| |
Collapse
|
8
|
Lenin R, Jha KA, Gentry J, Shrestha A, Culp EV, Vaithianathan T, Gangaraju R. Tauroursodeoxycholic Acid Alleviates Endoplasmic Reticulum Stress-Mediated Visual Deficits in Diabetic tie2-TNF Transgenic Mice via TGR5 Signaling. J Ocul Pharmacol Ther 2023; 39:159-174. [PMID: 36791327 PMCID: PMC10081728 DOI: 10.1089/jop.2022.0117] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 12/11/2022] [Indexed: 02/17/2023] Open
Abstract
Purpose: This study evaluated if tauroursodeoxycholic acid (TUDCA) alleviates pro-inflammatory and endoplasmic reticulum (ER) stress-mediated visual deficits in diabetic tie2-TNF transgenic mice via Takeda G protein-coupled receptor 5 (TGR5) receptor signaling. Methods: Adult tie2-TNF transgenic or age-matched C57BL/6J (wildtype, WT) mice were made diabetic and treated subcutaneously with TUDCA. After 4 weeks, visual function, vascular permeability, immunohistology, and molecular analyses were assessed. Human retinal endothelial cells (HRECs) silenced for TGR5, followed by TNF and high glucose (HG) stress-mediated endothelial permeability, and transendothelial migration of activated leukocytes were assessed with TUDCA in vitro. Results: Compared with WT mice, tie2-TNF mice showed a decreased visual function correlated with a decrease in protein kinase C α (PKCα) in rod bipolar cells, and increased vascular permeability was further exacerbated in diabetic-tie2-TNF mice. Conversely, TUDCA alleviated these changes in diabetic mice. An increase in inflammation and ER stress in retina coincided with an increase in TGR5 expression in diabetic tie2-TNF mice that decreased with TUDCA. In vitro, HRECs exposed to TNF+HG demonstrated >2-fold increase in TGR5 expression, a 3-fold increase in leukocyte transmigration with a concomitant increase in permeability. Although TUDCA reversed these effects, HRECs silenced for TGR5 and challenged with TUDCA or TGR5 agonist failed to reverse the TNF+HG induced effects. Conclusions: Our data suggest that TUDCA will serve as an excellent therapeutic agent for diabetic complications addressing both vascular and neurodegenerative changes in the retina. Perturbation of the TGR5 receptor in the retina might play a role in linking retinal ER stress to neurovascular dysfunction in diabetic retinopathy.
Collapse
Affiliation(s)
- Raji Lenin
- Department of Ophthalmology, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Kumar Abhiram Jha
- Department of Ophthalmology, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Jordy Gentry
- Department of Ophthalmology, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Abhishek Shrestha
- Department of Pharmacology, Addiction Science, and Toxicology, and The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Erielle V. Culp
- Department of Pharmacology, Addiction Science, and Toxicology, and The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Thirumalini Vaithianathan
- Department of Ophthalmology, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Department of Pharmacology, Addiction Science, and Toxicology, and The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Rajashekhar Gangaraju
- Department of Ophthalmology, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Department of Anatomy & Neurobiology, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
9
|
He Y, Yang M, Zhao R, Peng L, Dai E, Huang L, Zhao P, Li S, Yang Z. Novel truncating variants in CTNNB1 cause familial exudative vitreoretinopathy. J Med Genet 2023; 60:174-182. [PMID: 35361685 DOI: 10.1136/jmedgenet-2021-108259] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 03/12/2022] [Indexed: 01/27/2023]
Abstract
BACKGROUND Familial exudative vitreoretinopathy (FEVR) is an inheritable blinding disorder with clinical and genetic heterogeneity. Heterozygous variants in the CTNNB1 gene have been reported to cause FEVR. However, the pathogenic basis of CTNNB1-associated FEVR has not been fully explored. METHODS Whole-exome sequencing was performed on the genomic DNA of probands. Dual-luciferase reporter assay, western blotting and co-immunoprecipitation were used to characterise the impacts of variants. Quantitative real-time PCR, EdU (5-ethynyl-2'-deoxyuridine) incorporation assay and immunocytochemistry were performed on the primary human retinal microvascular endothelial cells (HRECs) to investigate the effect of CTNNB1 depletion on the downstream genes involved in Norrin/β-catenin signalling, cell proliferation and junctional integrity, respectively. Transendothelial electrical resistance assay was applied to measure endothelial permeability. Heterozygous endothelial-specific Ctnnb1-knockout mouse mice were generated to verify FEVR-like phenotypes in the retina. RESULTS We identified two novel heterozygous variants (p.Leu103Ter and p.Val199LeufsTer11) and one previously reported heterozygous variant (p.His369ThrfsTer2) in the CTNNB1 gene. These variants caused truncation and degradation of β-catenin that reduced Norrin/β-catenin signalling activity. Additionally, knockdown (KD) of CTNNB1 in HRECs led to diminished mRNA levels of Norrin/β-catenin targeted genes, reduced cell proliferation and compromised junctional integrity. The Cre-mediated heterozygous deletion of Ctnnb1 in mouse endothelial cells (ECs) resulted in FEVR-like phenotypes. Moreover, LiCl treatment partially rescued the defects in CTNNB1-KD HRECs and EC-specific Ctnnb1 heterozygous knockout mice. CONCLUSION Our findings reinforced the current pathogenesis of Norrin/β-catenin for FEVR and expanded the causative variant spectrum of CTNNB1 for the prenatal diagnosis and genetic counselling of FEVR.
Collapse
Affiliation(s)
- Yunqi He
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, Sichuan, China.,Sichuan Provincial Key Laboratory for Human Disease Gene Study, the Department of Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.,Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Mu Yang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, the Department of Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.,Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Rulian Zhao
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, the Department of Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.,Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Li Peng
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, the Department of Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.,Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Erkuan Dai
- Department of Ophthalmology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lulin Huang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, the Department of Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.,Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Peiquan Zhao
- Department of Ophthalmology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shujin Li
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, the Department of Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China .,Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Zhenglin Yang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, Sichuan, China .,Sichuan Provincial Key Laboratory for Human Disease Gene Study, the Department of Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.,Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
10
|
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
|
11
|
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
|
12
|
Seibold T, Schönfelder J, Weeber F, Lechel A, Armacki M, Waldenmaier M, Wille C, Palmer A, Halbgebauer R, Karasu E, Huber‐Lang M, Kalbitz M, Radermacher P, Paschke S, Seufferlein T, Eiseler T. Small Extracellular Vesicles Propagate the Inflammatory Response After Trauma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2102381. [PMID: 34713625 PMCID: PMC8693079 DOI: 10.1002/advs.202102381] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 08/20/2021] [Indexed: 05/03/2023]
Abstract
Trauma is the leading cause of death in individuals under 44 years of age. Thorax trauma (TxT) is strongly associated with trauma-related death, an unbalanced innate immune response, sepsis, acute respiratory distress syndrome, and multiple organ dysfunction. It is shown that different in vivo traumata, such as TxT or an in vitro polytrauma cytokine cocktail trigger secretion of small extracellular nanovesicles (sEVs) from endothelial cells with pro-inflammatory cargo. These sEVs transfer transcripts for ICAM-1, VCAM-1, E-selectin, and cytokines to systemically activate the endothelium, facilitate neutrophil-endothelium interactions, and destabilize barrier integrity. Inhibition of sEV-release after TxT in mice ameliorates local as well as systemic inflammation, neutrophil infiltration, and distant organ damage in kidneys (acute kidney injury, AKI). Vice versa, injection of TxT-plasma-sEVs into healthy animals is sufficient to trigger pulmonary and systemic inflammation as well as AKI. Accordingly, increased sEV concentrations and transfer of similar cargos are observed in polytrauma patients, suggesting a fundamental pathophysiological mechanism.
Collapse
Affiliation(s)
- Tanja Seibold
- Department of Internal Medicine IUniversity Hospital UlmAlbert‐Einstein‐Allee 23Ulm89081Germany
| | - Jonathan Schönfelder
- Department of Internal Medicine IUniversity Hospital UlmAlbert‐Einstein‐Allee 23Ulm89081Germany
| | - Florian Weeber
- Department of Internal Medicine IUniversity Hospital UlmAlbert‐Einstein‐Allee 23Ulm89081Germany
| | - André Lechel
- Department of Internal Medicine IUniversity Hospital UlmAlbert‐Einstein‐Allee 23Ulm89081Germany
| | - Milena Armacki
- Department of Internal Medicine IUniversity Hospital UlmAlbert‐Einstein‐Allee 23Ulm89081Germany
| | - Mareike Waldenmaier
- Department of Internal Medicine IUniversity Hospital UlmAlbert‐Einstein‐Allee 23Ulm89081Germany
| | - Christoph Wille
- Department of Internal Medicine IUniversity Hospital UlmAlbert‐Einstein‐Allee 23Ulm89081Germany
| | - Annette Palmer
- Institute of Clinical and Experimental Trauma‐ImmunologyUniversity Hospital UlmAlbert‐Einstein‐Allee 23Ulm89081Germany
| | - Rebecca Halbgebauer
- Institute of Clinical and Experimental Trauma‐ImmunologyUniversity Hospital UlmAlbert‐Einstein‐Allee 23Ulm89081Germany
| | - Ebru Karasu
- Institute of Clinical and Experimental Trauma‐ImmunologyUniversity Hospital UlmAlbert‐Einstein‐Allee 23Ulm89081Germany
| | - Markus Huber‐Lang
- Institute of Clinical and Experimental Trauma‐ImmunologyUniversity Hospital UlmAlbert‐Einstein‐Allee 23Ulm89081Germany
| | - Miriam Kalbitz
- Department of TraumatologyHandPlastic and Reconstructive SurgeryUniversity Hospital UlmAlbert‐Einstein‐Allee 23Ulm89081Germany
| | - Peter Radermacher
- Institute of Anesthesiological Pathophysiology and Process EngineeringUniversity Hospital UlmAlbert‐Einstein‐Allee 23Ulm89081Germany
| | - Stephan Paschke
- Department of General and Visceral SurgeryUniversity HospitalAlbert‐Einstein‐Allee 23Ulm89081Germany
| | - Thomas Seufferlein
- Department of Internal Medicine IUniversity Hospital UlmAlbert‐Einstein‐Allee 23Ulm89081Germany
| | - Tim Eiseler
- Department of Internal Medicine IUniversity Hospital UlmAlbert‐Einstein‐Allee 23Ulm89081Germany
| |
Collapse
|
13
|
Mishchenko EL, Mishchenko AM, Ivanisenko VA. Mechanosensitive molecular interactions in atherogenic regions of the arteries: development of atherosclerosis. Vavilovskii Zhurnal Genet Selektsii 2021; 25:552-561. [PMID: 34595377 PMCID: PMC8453358 DOI: 10.18699/vj21.062] [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: 01/27/2021] [Revised: 03/26/2021] [Accepted: 04/08/2021] [Indexed: 11/30/2022] Open
Abstract
A terrible disease of the cardiovascular system, atherosclerosis, develops in the areas of bends and
branches of arteries, where the direction and modulus of the blood flow velocity vector change, and consequently
so does the mechanical effect on endothelial cells in contact with the blood flow. The review focuses on topical
research studies on the development of atherosclerosis – mechanobiochemical events that transform the proatherogenic
mechanical stimulus of blood flow – low and low/oscillatory arterial wall shear stress in the chains of biochemical
reactions in endothelial cells, leading to the expression of specific proteins that cause the progression
of the pathological process. The stages of atherogenesis, systemic risk factors for atherogenesis and its important
hemodynamic factor, low and low/oscillatory wall shear stress exerted by blood flow on the endothelial cells lining
the arterial walls, have been described. The interactions of cell adhesion molecules responsible for the development
of atherosclerosis under low and low/oscillating shear stress conditions have been demonstrated. The activation
of the regulator of the expression of cell adhesion molecules, the transcription factor NF-κB, and the factors
regulating its activation under these conditions have been described. Mechanosensitive signaling pathways leading
to the expression of NF-κB in endothelial cells have been described. Studies of the mechanobiochemical signaling
pathways and interactions involved in the progression of atherosclerosis provide valuable information for the
development of approaches that delay or block the development of this disease.
Key words: atherogenesis; shear stress; transcription factor NF-κB; RelA expression; mechanosensitive receptors;
cell adhesion molecules; signaling pathways; mechanotransduction.
Collapse
Affiliation(s)
- E L Mishchenko
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | | | - V A Ivanisenko
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
14
|
Grimsley-Myers CM, Isaacson RH, Cadwell CM, Campos J, Hernandes MS, Myers KR, Seo T, Giang W, Griendling KK, Kowalczyk AP. VE-cadherin endocytosis controls vascular integrity and patterning during development. J Cell Biol 2021; 219:151601. [PMID: 32232465 PMCID: PMC7199849 DOI: 10.1083/jcb.201909081] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 01/10/2020] [Accepted: 02/28/2020] [Indexed: 12/18/2022] Open
Abstract
Tissue morphogenesis requires dynamic intercellular contacts that are subsequently stabilized as tissues mature. The mechanisms governing these competing adhesive properties are not fully understood. Using gain- and loss-of-function approaches, we tested the role of p120-catenin (p120) and VE-cadherin (VE-cad) endocytosis in vascular development using mouse mutants that exhibit increased (VE-cadGGG/GGG) or decreased (VE-cadDEE/DEE) internalization. VE-cadGGG/GGG mutant mice exhibited reduced VE-cad-p120 binding, reduced VE-cad levels, microvascular hemorrhaging, and decreased survival. By contrast, VE-cadDEE/DEE mutants exhibited normal vascular permeability but displayed microvascular patterning defects. Interestingly, VE-cadDEE/DEE mutant mice did not require endothelial p120, demonstrating that p120 is dispensable in the context of a stabilized cadherin. In vitro, VE-cadDEE mutant cells displayed defects in polarization and cell migration that were rescued by uncoupling VE-cadDEE from actin. These results indicate that cadherin endocytosis coordinates cell polarity and migration cues through actin remodeling. Collectively, our results indicate that regulated cadherin endocytosis is essential for both dynamic cell movements and establishment of stable tissue architecture.
Collapse
Affiliation(s)
| | - Robin H Isaacson
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA
| | - Chantel M Cadwell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA
| | - Jazmin Campos
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA
| | - Marina S Hernandes
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - Kenneth R Myers
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA
| | - Tadahiko Seo
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA
| | - William Giang
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA
| | - Kathy K Griendling
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - Andrew P Kowalczyk
- Department of Cell Biology, Department of Dermatology, and Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA
| |
Collapse
|
15
|
Huang SS, Cao S, Lu CE, Qin YB, Yang JP. Effects of nicorandil on p120 expression in the spinal cord and dorsal root ganglion of rats with chronic postsurgical pain. Mol Med Rep 2020; 22:4821-4827. [PMID: 33173987 PMCID: PMC7646919 DOI: 10.3892/mmr.2020.11546] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 08/21/2020] [Indexed: 01/25/2023] Open
Abstract
Chronic postsurgical pain (CPSP) has a high incidence, but the underlying mechanism is not well understood. Accumulating evidence has suggested that central sensitization is the main mechanism of pain. To study the role of p120 in CPSP, a skin/muscle incision and retraction (SMIR) model was established, and immunofluorescence staining and western blotting were performed to analyze the expression of p120 in the spinal cord and dorsal root ganglion (DRG). The results demonstrated that SMIR increased the expression of p120 in the DRG and the spinal cord compared with the naive group. Furthermore, it was demonstrated that p120 was mainly distributed in the glial fibrillary acidic protein-positive astrocytes in the spinal cord, and in the neurofilament 200-positive medium and large neurons in the DRG. Our previous studies have shown that adenosine triphosphate-sensitive potassium channel (KATP) agonists can reduce postoperative pain in rats. Therefore, the changes in p120 were observed in the DRG and spinal cord of rats following the intraperitoneal injection of nicorandil, a KATP agonist. It was demonstrated that nicorandil administration could relieve mechanical pain experienced following SMIR in rats, and decrease the expression of p120 in the DRG and spinal cord. The results revealed that p120 may contribute to the prophylactic analgesic effect of nicorandil, thus providing a novel insight into the mechanism of CPSP prevention.
Collapse
Affiliation(s)
- Sai-Sai Huang
- Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China
| | - Su Cao
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Cui E Lu
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Yi-Bin Qin
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Jian-Ping Yang
- Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China
| |
Collapse
|
16
|
Yanagisawa K, Konno M, Liu H, Irie S, Mizushima T, Mori M, Doki Y, Eguchi H, Matsusaki M, Ishii H. A Four-Dimensional Organoid System to Visualize Cancer Cell Vascular Invasion. BIOLOGY 2020; 9:biology9110361. [PMID: 33120912 PMCID: PMC7692192 DOI: 10.3390/biology9110361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 10/08/2020] [Accepted: 10/22/2020] [Indexed: 12/24/2022]
Abstract
Simple Summary Using vascular organoid culture with collagen microfiber, we have established a method for culturing organoids that recapitulates the vascular invasion process of cancer cells. This culture model made it possible to four-dimensionally evaluate the dynamics of cancer cells infiltrating into blood vessels. Abstract Vascular invasion of cancer is a critical step in cancer progression, but no drug has been developed to inhibit vascular invasion. To achieve the eradication of cancer metastasis, elucidation of the mechanism for vascular invasion and the development of innovative treatment methods are required. Here, a simple and reproducible vascular invasion model is established using a vascular organoid culture in a fibrin gel with collagen microfibers. Using this model, it was possible to observe and evaluate the cell dynamics and histological positional relationship of invasive cancer cells in four dimensions. Cancer-derived exosomes promoted the vascular invasion of cancer cells and loosened tight junctions in the vascular endothelium. As a new evaluation method, research using this vascular invasion mimic model will be advanced, and applications to the evaluation of the vascular invasion suppression effect of a drug are expected.
Collapse
Affiliation(s)
- Kiminori Yanagisawa
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan; (K.Y.); (M.K.); (T.M.); (M.M.); (Y.D.); (H.E.)
- Center of Molecular Innovation and Translational Research, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Masamitsu Konno
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan; (K.Y.); (M.K.); (T.M.); (M.M.); (Y.D.); (H.E.)
- Center of Molecular Innovation and Translational Research, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Hao Liu
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Suita, Osaka 565-0871, Japan;
| | - Shinji Irie
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduated School of Engineering, Osaka University, Suita, Osaka 565-0871, Japan;
| | - Tsunekazu Mizushima
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan; (K.Y.); (M.K.); (T.M.); (M.M.); (Y.D.); (H.E.)
| | - Masaki Mori
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan; (K.Y.); (M.K.); (T.M.); (M.M.); (Y.D.); (H.E.)
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 819-0395, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan; (K.Y.); (M.K.); (T.M.); (M.M.); (Y.D.); (H.E.)
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan; (K.Y.); (M.K.); (T.M.); (M.M.); (Y.D.); (H.E.)
| | - Michiya Matsusaki
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Suita, Osaka 565-0871, Japan;
- Correspondence: (M.M.); (H.I.); Tel.: +81-(0)6-6879-7357 (M.M.); +81-(0)6-6879-3251 or +81-(0)6-6210-8406 (H.I.); Fax: +81-(0)6-6879-7359 (M.M.); +81-(0)6-6879-3259 or +81-(0)6-4703-2856 (H.I.)
| | - Hideshi Ishii
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan; (K.Y.); (M.K.); (T.M.); (M.M.); (Y.D.); (H.E.)
- Center of Molecular Innovation and Translational Research, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
- Correspondence: (M.M.); (H.I.); Tel.: +81-(0)6-6879-7357 (M.M.); +81-(0)6-6879-3251 or +81-(0)6-6210-8406 (H.I.); Fax: +81-(0)6-6879-7359 (M.M.); +81-(0)6-6879-3259 or +81-(0)6-4703-2856 (H.I.)
| |
Collapse
|
17
|
Greig J, Bulgakova NA. Interplay between actomyosin and E-cadherin dynamics regulates cell shape in the Drosophila embryonic epidermis. J Cell Sci 2020; 133:jcs242321. [PMID: 32665321 DOI: 10.1242/jcs.242321] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 07/01/2020] [Indexed: 01/03/2023] Open
Abstract
Precise regulation of cell shape is vital for building functional tissues. Here, we study the mechanisms that lead to the formation of highly elongated anisotropic epithelial cells in the Drosophila epidermis. We demonstrate that this cell shape is the result of two counteracting mechanisms at the cell surface that regulate the degree of elongation: actomyosin, which inhibits cell elongation downstream of RhoA (Rho1 in Drosophila) and intercellular adhesion, modulated via clathrin-mediated endocytosis of E-cadherin (encoded by shotgun in flies), which promotes cell elongation downstream of the GTPase Arf1 (Arf79F in Drosophila). We show that these two mechanisms do not act independently but are interconnected, with RhoA signalling reducing Arf1 recruitment to the plasma membrane. Additionally, cell adhesion itself regulates both mechanisms - p120-catenin, a regulator of intercellular adhesion, promotes the activity of both Arf1 and RhoA. Altogether, we uncover a complex network of interactions between cell-cell adhesion, the endocytic machinery and the actomyosin cortex, and demonstrate how this network regulates cell shape in an epithelial tissue in vivo.
Collapse
Affiliation(s)
- Joshua Greig
- Department of Biomedical Science and Bateson Centre, The University of Sheffield, Sheffield S10 2TN, UK
| | - Natalia A Bulgakova
- Department of Biomedical Science and Bateson Centre, The University of Sheffield, Sheffield S10 2TN, UK
| |
Collapse
|
18
|
Unfractionated heparin attenuates endothelial barrier dysfunction via the phosphatidylinositol-3 kinase/serine/threonine kinase/nuclear factor kappa-B pathway. Chin Med J (Engl) 2020; 133:1815-1823. [PMID: 32649510 PMCID: PMC7470014 DOI: 10.1097/cm9.0000000000000905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Background Vascular endothelial dysfunction is considered a key pathophysiologic process for the development of acute lung injury. In this study, we aimed at investigating the effects of unfractionated heparin (UFH) on the lipopolysaccharide (LPS)-induced changes of vascular endothelial-cadherin (VE-cadherin) and the potential underlying mechanisms. Methods Male C57BL/6 J mice were randomized into three groups: vehicle, LPS, and LPS + UFH groups. Intraperitoneal injection of 30 mg/kg LPS was used to induce sepsis. Mice in the LPS + UFH group received subcutaneous injection of 8 U UFH 0.5 h before LPS injection. The lung tissue of the mice was collected for assessing lung injury by measuring the lung wet/dry (W/D) weight ratio and observing histological changes. Human pulmonary microvascular endothelial cells (HPMECs) were cultured and used to analyze the effects of UFH on LPS- or tumor necrosis factor-alpha (TNF-α)-induced vascular hyperpermeability, membrane expression of VE-cadherin, p120-catenin, and phosphorylated myosin light chain (p-MLC), and F-actin remodeling, and on the LPS-induced activation of the phosphatidylinositol-3 kinase (PI3K)/serine/threonine kinase (Akt)/nuclear factor kappa-B (NF-κB) signaling pathway. Results In vivo, UFH pretreatment significantly attenuated LPS-induced pulmonary histopathological changes (neutrophil infiltration and erythrocyte effusion, alveolus pulmonis collapse, and thicker septum), decreased the lung W/D, and increased protein concentration (LPS vs. LPS + UFH: 0.57 ± 0.04 vs. 0.32 ± 0.04 mg/mL, P = 0.0092), total cell count (LPS vs. LPS + UFH: 9.57 ± 1.23 vs. 3.65 ± 0.78 × 105/mL, P = 0.0155), polymorphonuclear neutrophil percentage (LPS vs. LPS + UFH: 88.05% ± 2.88% vs. 22.20% ± 3.92%, P = 0.0002), and TNF-α (460.33 ± 23.48 vs. 189.33 ± 14.19 pg/mL, P = 0.0006) in the bronchoalveolar lavage fluid. In vitro, UFH pre-treatment prevented the LPS-induced decrease in the membrane expression of VE-cadherin (LPS vs. LPS + UFH: 0.368 ± 0.044 vs. 0.716 ± 0.064, P = 0.0114) and p120-catenin (LPS vs. LPS + UFH: 0.208 ± 0.018 vs. 0.924 ± 0.092, P = 0.0016), and the LPS-induced increase in the expression of p-MLC (LPS vs. LPS + UFH: 0.972 ± 0.092 vs. 0.293 ± 0.025, P = 0.0021). Furthermore, UFH attenuated LPS- and TNF-α-induced hyperpermeability of HPMECs (LPS vs. LPS + UFH: 8.90 ± 0.66 vs. 15.84 ± 1.09 Ω·cm2, P = 0.0056; TNF-α vs. TNF-α + UFH: 11.28 ± 0.64 vs. 18.15 ± 0.98 Ω·cm2, P = 0.0042) and F-actin remodeling (LPS vs. LPS + UFH: 56.25 ± 1.51 vs. 39.70 ± 1.98, P = 0.0027; TNF-α vs. TNF-α + UFH: 55.42 ± 1.42 vs. 36.51 ± 1.20, P = 0.0005) in vitro. Additionally, UFH decreased the phosphorylation of Akt (LPS vs. LPS + UFH: 0.977 ± 0.081 vs. 0.466 ± 0.035, P = 0.0045) and I kappa B Kinase (IKK) (LPS vs. LPS + UFH: 1.023 ± 0.070 vs. 0.578 ± 0.044, P = 0.0060), and the nuclear translocation of NF-κB (LPS vs. LPS + UFH: 1.003 ± 0.077 vs. 0.503 ± 0.065, P = 0.0078) in HPMECs, which was similar to the effect of the PI3K inhibitor, wortmannin. Conclusions The protective effect of UFH against LPS-induced pulmonary endothelial barrier dysfunction involves VE-cadherin stabilization and PI3K/Akt/NF-κB signaling.
Collapse
|
19
|
Angé M, Castanares-Zapatero D, De Poortere J, Dufeys C, Courtoy GE, Bouzin C, Quarck R, Bertrand L, Beauloye C, Horman S. α1AMP-Activated Protein Kinase Protects against Lipopolysaccharide-Induced Endothelial Barrier Disruption via Junctional Reinforcement and Activation of the p38 MAPK/HSP27 Pathway. Int J Mol Sci 2020; 21:ijms21155581. [PMID: 32759774 PMCID: PMC7432762 DOI: 10.3390/ijms21155581] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/28/2020] [Accepted: 07/30/2020] [Indexed: 12/11/2022] Open
Abstract
Vascular hyperpermeability is a determinant factor in the pathophysiology of sepsis. While, AMP-activated protein kinase (AMPK) is known to play a role in maintaining endothelial barrier function in this condition. Therefore, we investigated the underlying molecular mechanisms of this protective effect. α1AMPK expression and/or activity was modulated in human dermal microvascular endothelial cells using either α1AMPK-targeting small interfering RNA or the direct pharmacological AMPK activator 991, prior to lipopolysaccharide (LPS) treatment. Western blotting was used to analyze the expression and/or phosphorylation of proteins that compose cellular junctions (zonula occludens-1 (ZO-1), vascular endothelial cadherin (VE-Cad), connexin 43 (Cx43)) or that regulate actin cytoskeleton (p38 MAPK; heat shock protein 27 (HSP27)). Functional endothelial permeability was assessed by in vitro Transwell assays, and quantification of cellular junctions in the plasma membrane was assessed by immunofluorescence. Actin cytoskeleton remodeling was evaluated through actin fluorescent staining. We consequently demonstrate that α1AMPK deficiency is associated with reduced expression of CX43, ZO-1, and VE-Cad, and that the drastic loss of CX43 is likely responsible for the subsequent decreased expression and localization of ZO-1 and VE-Cad in the plasma membrane. Moreover, α1AMPK activation by 991 protects against LPS-induced endothelial barrier disruption by reinforcing cortical actin cytoskeleton. This is due to a mechanism that involves the phosphorylation of p38 MAPK and HSP27, which is nonetheless independent of the small GTPase Rac1. This results in a drastic decrease of LPS-induced hyperpermeability. We conclude that α1AMPK activators that are suitable for clinical use may provide a specific therapeutic intervention that limits sepsis-induced vascular leakage.
Collapse
Affiliation(s)
- Marine Angé
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (M.A.); (D.C.-Z.); (J.D.P.); (C.D.); (L.B.); (C.B.)
| | - Diego Castanares-Zapatero
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (M.A.); (D.C.-Z.); (J.D.P.); (C.D.); (L.B.); (C.B.)
- Division of Intensive Care, Cliniques Universitaires Saint Luc, 1200 Brussels, Belgium
| | - Julien De Poortere
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (M.A.); (D.C.-Z.); (J.D.P.); (C.D.); (L.B.); (C.B.)
| | - Cécile Dufeys
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (M.A.); (D.C.-Z.); (J.D.P.); (C.D.); (L.B.); (C.B.)
| | - Guillaume E. Courtoy
- IREC Imaging Platform, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (G.E.C.); (C.B.)
| | - Caroline Bouzin
- IREC Imaging Platform, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (G.E.C.); (C.B.)
| | - Rozenn Quarck
- Department of Chronic Diseases & Metabolism (CHROMETA), Laboratory of Respiratory Diseases & Thoracic Surgery (BREATHE), KU Leuven, 3000 Leuven, Belgium;
| | - Luc Bertrand
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (M.A.); (D.C.-Z.); (J.D.P.); (C.D.); (L.B.); (C.B.)
| | - Christophe Beauloye
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (M.A.); (D.C.-Z.); (J.D.P.); (C.D.); (L.B.); (C.B.)
- Division of Cardiology, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Sandrine Horman
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (M.A.); (D.C.-Z.); (J.D.P.); (C.D.); (L.B.); (C.B.)
- Correspondence: ; Tel.: +32-2-764-55-66
| |
Collapse
|
20
|
Xu H, Pumiglia K, LaFlamme SE. Laminin-511 and α6 integrins regulate the expression of CXCR4 to promote endothelial morphogenesis. J Cell Sci 2020; 133:jcs246595. [PMID: 32409567 DOI: 10.1242/jcs.246595] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 04/11/2020] [Indexed: 12/25/2022] Open
Abstract
During angiogenesis, endothelial cells engage components of the extracellular matrix through integrin-mediated adhesion. Endothelial expression of laminin-411 and laminin-511 is known to promote vessel stability. However, little is known about the contribution of these laminins to endothelial morphogenesis. We used two organotypic cell culture angiogenesis assays, in conjunction with RNAi approaches, to demonstrate that depletion of either the α4 chain of laminin-411 (LAMA4) or the α5 chain of laminin-511 (LAMA5) from endothelial cells inhibits sprouting and tube formation. Depletion of α6 (ITGA6) integrins resulted in similar phenotypes. Gene expression analysis indicated that loss of either laminin-511 or α6 integrins inhibited the expression of CXCR4, a gene previously associated with angiogenic endothelial cells. Pharmacological or RNAi-dependent inhibition of CXCR4 suppressed endothelial sprouting and morphogenesis. Importantly, expression of recombinant CXCR4 rescued endothelial morphogenesis when α6 integrin expression was inhibited. Additionally, the depletion of α6 integrins from established tubes resulted in the loss of tube integrity and laminin-511. Taken together, our results indicate that α6 integrins and laminin-511 can promote endothelial morphogenesis by regulating the expression of CXCR4 and suggest that the α6-dependent deposition of laminin-511 protects the integrity of established endothelial tubes.
Collapse
Affiliation(s)
- Hao Xu
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany NY 12208, USA
| | - Kevin Pumiglia
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany NY 12208, USA
| | - Susan E LaFlamme
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany NY 12208, USA
| |
Collapse
|
21
|
Jinnouchi H, Guo L, Sakamoto A, Sato Y, Cornelissen A, Kawakami R, Mori M, Torii S, Kuntz S, Harari E, Mori H, Fuller D, Gadhoke N, Fernandez R, Paek KH, Surve D, Romero M, Kolodgie FD, Virmani R, Finn AV. Advances in mammalian target of rapamycin kinase inhibitors: application to devices used in the treatment of coronary artery disease. Future Med Chem 2020; 12:1181-1195. [PMID: 32431177 PMCID: PMC7333590 DOI: 10.4155/fmc-2019-0304] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 03/16/2020] [Indexed: 12/20/2022] Open
Abstract
Mammalian target of rapamycin (mTOR) inhibitors have been applied to vascular coronary devices to avoid neointimal growth and have become the predominant pharmacological agents used to prevent restenosis. mTOR inhibitors can affect not only proliferating vascular smooth muscle cells but also endothelial cells and therefore can result in delayed healing of the vessel including endothelialization. Emerging evidence suggests accelerated atherosclerosis due to the downstream negative effects on endothelial barrier functional recovery. The development of neoatherosclerosis within the neointima of drug-eluting stents can result in late thrombotic events. This type of problematic healing response may open the way for specific mTOR kinase inhibitors, such as ATP-competitive mTOR inhibitors. These inhibitors demonstrate a better healing profile than traditional limus-based drug-eluting stent and their clinical efficacy remains unknown.
Collapse
Affiliation(s)
- Hiroyuki Jinnouchi
- Cardiovascular Department, CVPath Institute, Gaithersburg, MD 20878, USA
| | - Liang Guo
- Cardiovascular Department, CVPath Institute, Gaithersburg, MD 20878, USA
| | - Atsushi Sakamoto
- Cardiovascular Department, CVPath Institute, Gaithersburg, MD 20878, USA
| | - Yu Sato
- Cardiovascular Department, CVPath Institute, Gaithersburg, MD 20878, USA
| | - Anne Cornelissen
- Cardiovascular Department, CVPath Institute, Gaithersburg, MD 20878, USA
| | - Rika Kawakami
- Cardiovascular Department, CVPath Institute, Gaithersburg, MD 20878, USA
| | - Masayuki Mori
- Cardiovascular Department, CVPath Institute, Gaithersburg, MD 20878, USA
| | - Sho Torii
- Cardiovascular Department, CVPath Institute, Gaithersburg, MD 20878, USA
| | - Salome Kuntz
- Cardiovascular Department, CVPath Institute, Gaithersburg, MD 20878, USA
| | - Emanuel Harari
- Cardiovascular Department, CVPath Institute, Gaithersburg, MD 20878, USA
| | - Hiroyoshi Mori
- Cardiovascular Department, CVPath Institute, Gaithersburg, MD 20878, USA
| | - Daniela Fuller
- Cardiovascular Department, CVPath Institute, Gaithersburg, MD 20878, USA
| | - Neel Gadhoke
- Cardiovascular Department, CVPath Institute, Gaithersburg, MD 20878, USA
| | - Raquel Fernandez
- Cardiovascular Department, CVPath Institute, Gaithersburg, MD 20878, USA
| | - Ka Hyun Paek
- Cardiovascular Department, CVPath Institute, Gaithersburg, MD 20878, USA
| | - Dipti Surve
- Cardiovascular Department, CVPath Institute, Gaithersburg, MD 20878, USA
| | - Maria Romero
- Cardiovascular Department, CVPath Institute, Gaithersburg, MD 20878, USA
| | - Frank D Kolodgie
- Cardiovascular Department, CVPath Institute, Gaithersburg, MD 20878, USA
| | - Renu Virmani
- Cardiovascular Department, CVPath Institute, Gaithersburg, MD 20878, USA
| | - Aloke V Finn
- Cardiovascular Department, CVPath Institute, Gaithersburg, MD 20878, USA
| |
Collapse
|
22
|
Smith RO, Ninchoji T, Gordon E, André H, Dejana E, Vestweber D, Kvanta A, Claesson-Welsh L. Vascular permeability in retinopathy is regulated by VEGFR2 Y949 signaling to VE-cadherin. eLife 2020; 9:54056. [PMID: 32312382 PMCID: PMC7188482 DOI: 10.7554/elife.54056] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 04/05/2020] [Indexed: 01/01/2023] Open
Abstract
Edema stemming from leaky blood vessels is common in eye diseases such as age-related macular degeneration and diabetic retinopathy. Whereas therapies targeting vascular endothelial growth factor A (VEGFA) can suppress leakage, side-effects include vascular rarefaction and geographic atrophy. By challenging mouse models representing different steps in VEGFA/VEGF receptor 2 (VEGFR2)-induced vascular permeability, we show that targeting signaling downstream of VEGFR2 pY949 limits vascular permeability in retinopathy induced by high oxygen or by laser-wounding. Although suppressed permeability is accompanied by reduced pathological neoangiogenesis in oxygen-induced retinopathy, similarly sized lesions leak less in mutant mice, separating regulation of permeability from angiogenesis. Strikingly, vascular endothelial (VE)-cadherin phosphorylation at the Y685, but not Y658, residue is reduced when VEGFR2 pY949 signaling is impaired. These findings support a mechanism whereby VE-cadherin Y685 phosphorylation is selectively associated with excessive vascular leakage. Therapeutically, targeting VEGFR2-regulated VE-cadherin phosphorylation could suppress edema while leaving other VEGFR2-dependent functions intact. The number of people with impaired vision and blindness is increasing in Western society due to the aging population and the increased prevalence of diabetes. This has led to eye diseases, such as age-related macular degeneration and diabetic retinopathy becoming more common. In both these eye diseases, new blood vessels grow in the retina – the light-sensitive part of the eye – to bring oxygen and nutrients to the tissue. However, these new blood vessels are leaky and allow molecules to leave the bloodstream and enter the retinal tissue. This causes the retina to swell and impair a person’s vision. The leaky blood supply also reduces the amount of oxygen that gets to the tissue, resulting in further damage to the retina. When tissues experience low levels of oxygen, cells start making a protein called vascular endothelial growth factor (or VEGF for short). Whilst this protein is important for helping form new blood vessels, it also makes these vessels leaky. Current treatments for age-related macular degeneration and diabetic retinopathy decrease swelling in the eye by blocking the action of VEGF. However, these treatments also cause existing blood vessels and nerve cells to die, leading to irreversible damage. Now, Smith et al. have set out to find whether the effects of VEGF can be blocked without causing further damage to existing cells. To investigate this possibility, the eyes and retinas of mice were treated with a laser or exposed to changing oxygen levels to create injuries that resembled human age-related macular degeneration and diabetic retinopathy. Each of the tested mice had specific mutations in proteins known to interact with VEGF. Fluorescent particles were injected into the bloodstream of the mice to assess how these different mutations affected blood vessel leakage: if fluorescent particles could no longer be detected outside the blood vessels, this suggested that the mutation had stopped the vessels from leaking. Further experiments showed these specific mutations affected leakage and did not prevent new blood vessels from forming. In the future it will be important to see if drugs, rather than mutations, can also decrease the leakiness of blood vessels in the retina. Such chemical compounds could then be tested in mouse experiments. If successful, these drugs might be used to treat patients with age-related macular degeneration and diabetic retinopathy.
Collapse
Affiliation(s)
- Ross O Smith
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Science for Life Laboratory Uppsala University, Uppsala, Sweden
| | - Takeshi Ninchoji
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Science for Life Laboratory Uppsala University, Uppsala, Sweden
| | - Emma Gordon
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Science for Life Laboratory Uppsala University, Uppsala, Sweden
| | - Helder André
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Elisabetta Dejana
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Science for Life Laboratory Uppsala University, Uppsala, Sweden.,IFOM-IEO Campus Via Adamello, Milan, Italy
| | | | - Anders Kvanta
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Lena Claesson-Welsh
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Science for Life Laboratory Uppsala University, Uppsala, Sweden
| |
Collapse
|
23
|
GRP78 translocation to the cell surface and O-GlcNAcylation of VE-Cadherin contribute to ER stress-mediated endothelial permeability. Sci Rep 2019; 9:10783. [PMID: 31346222 PMCID: PMC6658495 DOI: 10.1038/s41598-019-47246-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 07/10/2019] [Indexed: 12/31/2022] Open
Abstract
Increased O-GlcNAcylation, a well-known post-translational modification of proteins causally linked to various detrimental cellular functions in pathological conditions including diabetic retinopathy (DR). Previously we have shown that endothelial activation induced by inflammation and hyperglycemia results in the endoplasmic reticulum (ER) stress-mediated intercellular junction alterations accompanied by visual deficits in a tie2-TNF-α transgenic mouse model. In this study, we tested the hypothesis that increased ER stress via O-GlcNAcylation of VE-Cadherin likely contribute to endothelial permeability. We show that ER stress leads to GRP78 translocation to the plasma membrane, increased O-GlcNAcylation of proteins, particularly VE-Cadherin resulting in a defective complex partnering leading to the loss of retinal endothelial barrier integrity and increased transendothelial migration of monocytes. We further show an association of GRP78 with the VE-Cadherin under these conditions. Interestingly, cells exposed to ER stress inhibitor, tauroursodeoxycholic acid partially mitigated all these effects. Our findings suggest an essential role for ER stress and O-GlcNAcylation in altering the endothelial barrier function and reveal a potential therapeutic target in the treatment of DR.
Collapse
|
24
|
Different concentrations of lipopolysaccharide regulate barrier function through the PI3K/Akt signalling pathway in human pulmonary microvascular endothelial cells. Sci Rep 2018; 8:9963. [PMID: 29967433 PMCID: PMC6028427 DOI: 10.1038/s41598-018-28089-3] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 06/14/2018] [Indexed: 12/15/2022] Open
Abstract
Lipopolysaccharide (LPS) can lead to vascular endothelial barrier dysfunction, which often results in acute lung injury and acute respiratory distress syndrome. However, the effects of different concentrations of LPS on human pulmonary microvascular endothelial barrier function and the involvement of the phosphatidylinositol-3-kinase-serine/threonine kinase (PI3K/Akt) pathway in this process remain unclear. Human pulmonary microvascular endothelial cells (HPMECs) were stimulated with different doses of LPS, and barrier function was examined by determining cell monolayer permeability, cell migration, and the expression of intercellular junction proteins (VE-Cadherin, Claudin-5, and Connexin-43). LY294002 was used to inhibit PI3K to verify the role of the PI3K/Akt pathway in the regulation of barrier function in HPMECs stimulated by LPS. Low doses of LPS increased HPMEC migration, up-regulated VE-Cadherin and Claudin-5 expression, down-regulated Connexin-43 expression, and promoted Akt phosphorylation, which could collectively decrease monolayer permeability. In contrast, high doses of LPS suppressed HPMEC migration, down-regulated the expression of VE-Cadherin and Claudin-5, up-regulated Connexin-43 expression, and reduced Akt phosphorylation, which could collectively increase monolayer permeability. LPS has a biphasic effect on HPMEC barrier function through the PI3K/Akt pathway, and this effect is concentration-dependent.
Collapse
|
25
|
Yang D, Qiu J, Xu N, Zhao Y, Li T, Ma Q, Huang J, Wang G. Mussel adhesive protein fused with VE-cadherin domain specifically triggers endothelial cell adhesion. J Mater Chem B 2018; 6:4151-4163. [PMID: 32255158 DOI: 10.1039/c8tb00526e] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Endothelium is the only known completely non-thrombogenic material. In the present study, a strategy to mimic the adhesive interactions of endothelial cells (ECs) to alter the vascular microenvironment was established and applied to directing the behaviour of cells. To facilitate the regeneration of a functional endothelium in vascular lesions, we designed a recombinant mussel foot protein (Mfp-5) fused with the VE-cadherin extracellular domain EC1-2, termed VE-M. Surface coating analysis showed that recombinant VE-M successfully formed a coating on substrate materials with uniform nanorods, low roughness, and sufficient hydrophilicity. We then evaluated the effects of VE-M on the adhesion of ECs and the capture of endothelial progenitor cells (EPCs). The result demonstrated that VE-M efficiently promoted the adhesion of ECs and EPCs. The number of ECs and EPCs on VE-M was 5.5- and 1.8-fold higher, respectively, than that on bare 316L SS under static conditions, whereas there was no significant difference in the number of captured smooth muscle cells (SMCs) between VE-M and other substrates. In addition, the number of EPCs captured by VE-M was approximately four times higher than that captured by 316L SS under dynamic conditions. In particular, the result of the neutralization test indicated that VE-M specifically triggered ECs' adhesion via the interaction of VE-cadherin EC1-2. Further investigation showed that VE-M significantly increased the levels of endogenous VE-cadherin in HUVECs as well as the endothelial eNOS content, with little or no endothelial inflammation. Our results showed that VE-M could be a promising biomimetic modification for accelerating endothelialization and vascularization in tissue engineering.
Collapse
Affiliation(s)
- Dongchuan Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400030, P. R. China.
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Alsaffar H, Martino N, Garrett JP, Adam AP. Interleukin-6 promotes a sustained loss of endothelial barrier function via Janus kinase-mediated STAT3 phosphorylation and de novo protein synthesis. Am J Physiol Cell Physiol 2018; 314:C589-C602. [PMID: 29351406 DOI: 10.1152/ajpcell.00235.2017] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Vascular leakage is a hallmark of the inflammatory response. Acute changes in endothelial permeability are due to posttranslational changes in intercellular adhesion and cytoskeleton proteins. However, little is known about the mechanisms leading to long-term changes in vascular permeability. Here, we show that interleukin-6 (IL-6) promotes an increase in endothelial monolayer permeability that lasts over 24 h and demonstrate that activation of Src and MEK/ERK pathways is required only for short-term increases in permeability, being dispensable after 2 h. In contrast, Janus kinase (JAK)-mediated STAT3 phosphorylation at Y705 (but not S727) and de novo synthesis of RNA and proteins are required for the sustained permeability increases. Loss of junctional localization of VE-cadherin and ZO-1 is evident several hours after the maximal IL-6 response, thus suggesting that these events are a consequence of IL-6 signaling, but not a cause of the increased permeability. Understanding the mechanisms involved in sustaining vascular permeability may prove crucial to allow us to directly target vascular leakage and minimize tissue damage, thus reducing the rates of mortality and chronic sequelae of excessive edema. Targeting endothelial-specific mechanisms regulating barrier function could provide a new therapeutic strategy to prevent vascular leakage while maintaining the immune response and other beneficial aspects of the inflammatory response that are required for bacterial clearance and tissue repair.
Collapse
Affiliation(s)
- Hiba Alsaffar
- Department of Molecular and Cellular Physiology, Albany Medical Center , Albany, New York
| | - Nina Martino
- Department of Molecular and Cellular Physiology, Albany Medical Center , Albany, New York
| | - Joshua P Garrett
- Department of Molecular and Cellular Physiology, Albany Medical Center , Albany, New York
| | - Alejandro P Adam
- Department of Molecular and Cellular Physiology, Albany Medical Center , Albany, New York.,Department of Ophthalmology, Albany Medical Center, Albany, New York
| |
Collapse
|