1
|
Vu HH, Moellmer SA, McCarty OJ, Puy C. New mechanisms and therapeutic approaches to regulate vascular permeability in systemic inflammation. Curr Opin Hematol 2025; 32:130-137. [PMID: 40063579 PMCID: PMC11949701 DOI: 10.1097/moh.0000000000000864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
PURPOSE OF REVIEW This review summarizes mechanisms that regulate endothelial vascular permeability in health and disease. In systemic inflammation, the endothelial barrier integrity is disrupted, which exacerbates vascular permeability, leading to organ failure and death. Herein we provide an overview of emerging therapeutic targets to reverse barrier dysfunction and preserve vascular permeability in inflammatory diseases like sepsis. RECENT FINDINGS Endothelial barrier function is regulated in part by the endothelial cell-specific protein, Roundabout 4 (ROBO4), and vascular endothelial (VE)-cadherin, a critical adherens junction protein, which act in concert to suppresses vascular permeability by stabilizing endothelial cell-cell interactions. We recently discovered a pathway by which activation of coagulation factor XI (FXI) enhances the cleavage of VE-cadherin by the metalloproteinase ADAM10, contributing to sepsis-related endothelial damage and loss of barrier function. Targeting FXI improved survival and reduced sVE-cadherin levels in a baboon model of sepsis while enhancing Robo4 expression decreased mortality in LPS-treated mice. SUMMARY Endothelial cell barrier dysfunction is a hallmark of excessive immune responses characteristic of systemic inflammatory diseases such as sepsis. Advances in understanding the molecular mechanisms regulating vascular permeability, for instance the newly discovered roles of FXI or ROBO4, may help identify novel therapeutic targets for mitigating vascular hyperpermeability in septic patients.
Collapse
Affiliation(s)
- Helen H. Vu
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | - Samantha A. Moellmer
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | - Owen J.T. McCarty
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
- Division of Hematology and Medical Oncology, Oregon Health and Science University, Portland, OR, USA
| | - Cristina Puy
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
2
|
Karki P, Ke Y, Zhang C, Promnares K, Li Y, Williams CH, Hong CC, Birukov KG, Birukova AA. Inhibition of proton sensor GPR68 suppresses endothelial dysfunction and acute lung injury caused by Staphylococcus aureus bacterial particles. FASEB J 2025; 39:e70333. [PMID: 39907683 PMCID: PMC11797537 DOI: 10.1096/fj.202401947r] [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: 08/19/2024] [Revised: 12/21/2024] [Accepted: 01/08/2025] [Indexed: 02/06/2025]
Abstract
Lung bacterial infections, including hospital-acquired pneumonia, remain a serious problem for public health. Endothelial cell (EC) exposure to heat-killed Staphylococcus aureus (HKSA) represents a clinical scenario of high titers of killed bacterial particles present in the host after antibiotic therapy, which triggers inflammatory cascades, cytokine storms, and EC dysfunction leading to acute lung injury (ALI). GPR68 is a member of the proton-sensing G protein-coupled receptor family. Acting as a pH sensor, GPR68 becomes activated upon pH reduction and contributes to pathologic cell responses by activating ER stress and unfolded protein response. This study investigated the role of GPR68 in HKSA-induced EC dysfunction and HKSA-induced ALI. HKSA robustly increased GPR68 mRNA levels in human pulmonary EC and directly stimulated GPR68 activity. A selective GPR68 small molecule inhibitor, OGM-8345, attenuated HKSA-induced EC permeability and protected cell junction integrity. OGM-8345 inhibited HKSA-induced activation of inflammatory genes TNF-α, IL-6, IL-8, IL-1β, and CXCL5 and decreased cytokine secretion by HKSA-challenged EC. Co-treatment with the GPR68 activator Ogerin or medium acidification to pH 6.5 augmented HKSA-induced EC dysfunction, which was rescued by OGM-8345. Intratracheal HKSA injection increased vascular leak and lung inflammation in mice which were monitored by lung Evans blue extravasation, increased cell and protein count in bronchoalveolar lavage, and mRNA expression of inflammatory genes. ALI and barrier dysfunction was attenuated by OGM-8345. We show for the first time the role of GPR68 in mediating HKSA-induced lung injury and the strong potential for OGM-8345 as a therapeutic treatment of bacterial pathogen-induced ALI associated with tissue acidification.
Collapse
Affiliation(s)
- Pratap Karki
- Division of Pulmonary and Critical Care, Department of MedicineUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Yunbo Ke
- Department of AnesthesiologyUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Chen‐Ou Zhang
- Division of Pulmonary and Critical Care, Department of MedicineUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Kamoltip Promnares
- Department of AnesthesiologyUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Yue Li
- Division of Pulmonary and Critical Care, Department of MedicineUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Charles H. Williams
- Division of Cardiovascular Medicine, Department of MedicineUniversity of Maryland School of MedicineBaltimoreMarylandUSA
- Present address:
Department of MedicineMichigan State University College of Human MedicineEast LansingMichiganUSA
| | - Charles C. Hong
- Division of Cardiovascular Medicine, Department of MedicineUniversity of Maryland School of MedicineBaltimoreMarylandUSA
- Present address:
Department of MedicineMichigan State University College of Human MedicineEast LansingMichiganUSA
| | - Konstantin G. Birukov
- Department of AnesthesiologyUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Anna A. Birukova
- Division of Pulmonary and Critical Care, Department of MedicineUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
3
|
Karki P, Ke Y, Zhang C, Promnares K, Li Y, Williams CH, Hong CC, Birukov KG, Birukova AA. GPR68 Mediates Lung Endothelial Dysfunction Caused by Bacterial Inflammation and Tissue Acidification. Cells 2024; 13:2125. [PMID: 39768215 PMCID: PMC11674861 DOI: 10.3390/cells13242125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 12/18/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
Tissue acidification resulting from dysregulated cellular bioenergetics accompanies various inflammatory states. GPR68, along with other members of proton-sensing G protein-coupled receptors, responds to extracellular acidification and has been implicated in chronic inflammation-related diseases such as ischemia, cancer, and colitis. The present study examined the role of extracellular acidification on human pulmonary endothelial cell (EC) permeability and inflammatory status per se and investigated potential synergistic effects of acidosis on endothelial dysfunction caused by bacterial lipopolysaccharide (LPS, Klebsiella pneumoniae). Results showed that medium acidification to pH 6.5 caused a delayed increase in EC permeability illustrated by a decrease in transendothelial electrical resistance and loss of continuous VE-cadherin immunostaining at cell junctions. Likewise, acidic pH induced endothelial inflammation reflected by increased mRNA and protein expression of EC adhesion molecules VCAM-1 and ICAM-1, upregulated mRNA transcripts of tumor necrosis factor-α, IL-6, IL-8, IL-1β, and CXCL5, and increased secretion of ICAM-1, IL-6, and IL-8 in culture medium monitored by ELISA. Among the GPCRs tested, acidic pH selectively increased mRNA and protein expression of GPR68, and only the GPR68-specific small molecule inhibitor OGM-8345 rescued acidosis-induced endothelial permeability and inflammation. Furthermore, acidic pH exacerbated LPS-induced endothelial permeability and inflammatory response in cultured lung macrovascular as well as microvascular endothelial cells. These effects were suppressed by OGM-8345 in both EC types. Altogether, these results suggest that GPR68 is a critical mediator of acidic pH-induced dysfunction of human pulmonary vascular endothelial cells and mediates the augmenting effect of tissue acidification on LPS-induced endothelial cell injury.
Collapse
Affiliation(s)
- Pratap Karki
- Division of Pulmonary and Critical Care, Department of Medicine, UMSOM Lung Biology Program, University of Maryland School of Medicine, 20 Penn Street, HSF-2, Room S143, Baltimore, MD 21201, USA; (P.K.); (C.Z.); (Y.L.)
| | - Yunbo Ke
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (Y.K.); (K.P.); (K.G.B.)
| | - Chenou Zhang
- Division of Pulmonary and Critical Care, Department of Medicine, UMSOM Lung Biology Program, University of Maryland School of Medicine, 20 Penn Street, HSF-2, Room S143, Baltimore, MD 21201, USA; (P.K.); (C.Z.); (Y.L.)
| | - Kamoltip Promnares
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (Y.K.); (K.P.); (K.G.B.)
| | - Yue Li
- Division of Pulmonary and Critical Care, Department of Medicine, UMSOM Lung Biology Program, University of Maryland School of Medicine, 20 Penn Street, HSF-2, Room S143, Baltimore, MD 21201, USA; (P.K.); (C.Z.); (Y.L.)
| | - Charles H. Williams
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (C.H.W.); (C.C.H.)
| | - Charles C. Hong
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (C.H.W.); (C.C.H.)
| | - Konstantin G. Birukov
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (Y.K.); (K.P.); (K.G.B.)
| | - Anna A. Birukova
- Division of Pulmonary and Critical Care, Department of Medicine, UMSOM Lung Biology Program, University of Maryland School of Medicine, 20 Penn Street, HSF-2, Room S143, Baltimore, MD 21201, USA; (P.K.); (C.Z.); (Y.L.)
| |
Collapse
|
4
|
Stefanenko M, Fedoriuk M, Mamenko M, Semenikhina M, Nowling TK, Lipschutz JH, Maximyuk O, Staruschenko A, Palygin O. PAR1-mediated Non-periodical Synchronized Calcium Oscillations in Human Mesangial Cells. FUNCTION 2024; 5:zqae030. [PMID: 38984988 PMCID: PMC11384906 DOI: 10.1093/function/zqae030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 07/11/2024] Open
Abstract
Mesangial cells offer structural support to the glomerular tuft and regulate glomerular capillary flow through their contractile capabilities. These cells undergo phenotypic changes, such as proliferation and mesangial expansion, resulting in abnormal glomerular tuft formation and reduced capillary loops. Such adaptation to the changing environment is commonly associated with various glomerular diseases, including diabetic nephropathy and glomerulonephritis. Thrombin-induced mesangial remodeling was found in diabetic patients, and expression of the corresponding protease-activated receptors (PARs) in the renal mesangium was reported. However, the functional PAR-mediated signaling in mesangial cells was not examined. This study investigated protease-activated mechanisms regulating mesangial cell calcium waves that may play an essential role in the mesangial proliferation or constriction of the arteriolar cells. Our results indicate that coagulation proteases such as thrombin induce synchronized oscillations in cytoplasmic Ca2+ concentration of mesangial cells. The oscillations required PAR1 G-protein coupled receptors-related activation, but not a PAR4, and were further mediated presumably through store-operated calcium entry and transient receptor potential canonical 3 (TRPC3) channel activity. Understanding thrombin signaling pathways and their relation to mesangial cells, contractile or synthetic (proliferative) phenotype may play a role in the development of chronic kidney disease and requires further investigation.
Collapse
Affiliation(s)
- Mariia Stefanenko
- Department of Medicine, Division of Nephrology, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Cellular Membranology, Bogomoletz Institute of Physiology, Kyiv 01024, Ukraine
| | - Mykhailo Fedoriuk
- Department of Medicine, Division of Nephrology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Mykola Mamenko
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Marharyta Semenikhina
- Department of Medicine, Division of Nephrology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Tamara K Nowling
- Department of Medicine, Division of Rheumatology & Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Joshua H Lipschutz
- Department of Medicine, Division of Nephrology, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Medicine, Ralph H. Johnson VAMC, Charleston, SC 29401, USA
| | - Oleksandr Maximyuk
- Department of Cellular Membranology, Bogomoletz Institute of Physiology, Kyiv 01024, Ukraine
| | - Alexander Staruschenko
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL 33602, USA
- James A. Haley Veterans’ Hospital, Tampa, FL 33612, USA
| | - Oleg Palygin
- Department of Medicine, Division of Nephrology, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
5
|
Kim SY, Cheon J. Senescence-associated microvascular endothelial dysfunction: A focus on the blood-brain and blood-retinal barriers. Ageing Res Rev 2024; 100:102446. [PMID: 39111407 DOI: 10.1016/j.arr.2024.102446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 07/05/2024] [Accepted: 08/02/2024] [Indexed: 08/17/2024]
Abstract
The blood-brain barrier (BBB) and blood-retinal barrier (BRB) constitute critical physiochemical interfaces, precisely orchestrating the bidirectional communication between the brain/retina and blood. Increased permeability or leakage of these barriers has been demonstrably linked to age-related vascular and parenchymal damage. While it has been suggested that the gradual aging process may coincide with disruptions in these barriers, this phenomenon is significantly exacerbated in individuals with age-related neurodegenerative disorders (ARND). This review focuses on the microvascular endothelium, a key constituent of BBB and BRB, highlighting the impact of endothelial senescence on barrier dysfunction and exploring recent discoveries regarding core pathways implicated in its breakdown. Subsequently, we address the "vascular senescence hypothesis" for ARND, with a particular emphasis on Alzheimer's disease and age-related macular degeneration, centered on endothelial senescence. Finally, we discuss potential senotherapeutic strategies targeting barrier dysfunction.
Collapse
Affiliation(s)
- Sung Young Kim
- Department of Biochemistry, Konkuk University School of Medicine, Republic of Korea; Research Institute of Medical Science, Konkuk University, Republic of Korea; IBST, Konkuk University, Republic of Korea.
| | - Jaejoung Cheon
- Department of Biochemistry, Konkuk University School of Medicine, Republic of Korea
| |
Collapse
|
6
|
Wakasugi R, Suzuki K, Kaneko-Kawano T. Molecular Mechanisms Regulating Vascular Endothelial Permeability. Int J Mol Sci 2024; 25:6415. [PMID: 38928121 PMCID: PMC11203514 DOI: 10.3390/ijms25126415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/30/2024] [Accepted: 06/02/2024] [Indexed: 06/28/2024] Open
Abstract
Vascular endothelial cells form a monolayer in the vascular lumen and act as a selective barrier to control the permeability between blood and tissues. To maintain homeostasis, the endothelial barrier function must be strictly integrated. During acute inflammation, vascular permeability temporarily increases, allowing intravascular fluid, cells, and other components to permeate tissues. Moreover, it has been suggested that the dysregulation of endothelial cell permeability may cause several diseases, including edema, cancer, and atherosclerosis. Here, we reviewed the molecular mechanisms by which endothelial cells regulate the barrier function and physiological permeability.
Collapse
Affiliation(s)
| | | | - Takako Kaneko-Kawano
- Graduate School of Pharmacy, Ritsumeikan University, 1-1-1 Noji-higashi, Kusatsu 525-8577, Shiga, Japan; (R.W.); (K.S.)
| |
Collapse
|
7
|
Su Y, Lucas R, Fulton DJ, Verin AD. Mechanisms of pulmonary endothelial barrier dysfunction in acute lung injury and acute respiratory distress syndrome. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2024; 2:80-87. [PMID: 39006829 PMCID: PMC11242916 DOI: 10.1016/j.pccm.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Indexed: 07/16/2024]
Abstract
Endothelial cells (ECs) form a semi-permeable barrier between the interior space of blood vessels and the underlying tissues. Pulmonary endothelial barrier integrity is maintained through coordinated cellular processes involving receptors, signaling molecules, junctional complexes, and protein-regulated cytoskeletal reorganization. In acute lung injury (ALI) or its more severe form acute respiratory distress syndrome (ARDS), the loss of endothelial barrier integrity secondary to endothelial dysfunction caused by severe pulmonary inflammation and/or infection leads to pulmonary edema and hypoxemia. Pro-inflammatory agonists such as histamine, thrombin, bradykinin, interleukin 1β, tumor necrosis factor α, vascular endothelial growth factor, angiopoietin-2, and platelet-activating factor, as well as bacterial toxins and reactive oxygen species, cause dynamic changes in cytoskeletal structure, adherens junction disorganization, and detachment of vascular endothelial cadherin (VE-cadherin) from the actin cytoskeleton, leading to an increase in endothelial permeability. Endothelial interactions with leukocytes, platelets, and coagulation enhance the inflammatory response. Moreover, inflammatory infiltration and the associated generation of pro-inflammatory cytokines during infection cause EC death, resulting in further compromise of the structural integrity of lung endothelial barrier. Despite the use of potent antibiotics and aggressive intensive care support, the mortality of ALI is still high, because the mechanisms of pulmonary EC barrier disruption are not fully understood. In this review, we summarized recent advances in the studies of endothelial cytoskeletal reorganization, inter-endothelial junctions, endothelial inflammation, EC death, and endothelial repair in ALI and ARDS, intending to shed some light on the potential diagnostic and therapeutic targets in the clinical management of the disease.
Collapse
Affiliation(s)
- Yunchao Su
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Research Service, Charlie Norwood Veterans Affairs Medical Center, Augusta, GA 30912, USA
| | - Rudolf Lucas
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - David J.R. Fulton
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Alexander D. Verin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
8
|
Shi Y, Li D, Yi B, Tang H, Xu T, Zhang Y. Physiological cyclic stretching potentiates the cell-cell junctions in vascular endothelial layer formed on aligned fiber substrate. BIOMATERIALS ADVANCES 2024; 157:213751. [PMID: 38219418 DOI: 10.1016/j.bioadv.2023.213751] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 12/11/2023] [Accepted: 12/21/2023] [Indexed: 01/16/2024]
Abstract
In vascular tissue engineering, formation of stable endothelial cell-cell and cell-substrate adhesions is essential for maintaining long-term patency of the tissue-engineered vascular grafts (TEVGs). In this study, sheet-like aligned fibrous substrates of poly(l-lactide-co-caprolactone) (PLCL) were prepared by electrospinning to provide basement membrane-resembling structural support to endothelial cells (ECs). Cyclic stretching at physiological and pathological levels was then applied to human umbilical vein endothelial cells (HUVECs) cultured on chosen fibrous substrate using a force-loading device, from which effects of the cyclic stretching on cell-cell and cell-substrate adhesions were examined. It was found that applying uniaxial 1 Hz cyclic stretch at physiological levels (5 % and 10 % elongation) strengthened the cell-cell junctions, thus leading to improved structural integrity, functional expression and resistance to thrombin-induced damaging impacts in the formed endothelial layer. The cell-cell junctions were disrupted at pathological level (15 % elongation) cyclic stretching, which however facilitated the formation of focal adhesions (FAs) at cell-substrate interface. Mechanistically, the effects of cyclic stretching on endothelial cell-cell and cell-substrate adhesions were identified to be correlated with the RhoA/ROCK signaling pathway. Results from this study highlight the relevance between applying dynamic mechanical stimulation and maintaining the structural integrity of the formed endothelial layer, and implicate a necessity to implement appropriate dynamic mechanical training (i.e., preconditioning) to obtain tissue-engineered blood vessels with long-term patency post-implantation.
Collapse
Affiliation(s)
- Yu Shi
- College of Biological Science and Medical Engineering, Donghua University, Shanghai, China
| | - Donghong Li
- College of Biological Science and Medical Engineering, Donghua University, Shanghai, China
| | - Bingcheng Yi
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Han Tang
- College of Biological Science and Medical Engineering, Donghua University, Shanghai, China
| | - Tingting Xu
- College of Biological Science and Medical Engineering, Donghua University, Shanghai, China
| | - Yanzhong Zhang
- College of Biological Science and Medical Engineering, Donghua University, Shanghai, China; Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai, China.
| |
Collapse
|
9
|
Karki P, Li Y, Zhang CO, Ke Y, Promnares K, Birukova AA, Eggerman TL, Bocharov AV, Birukov KG. Amphipathic Helical Peptide L37pA Protects against Lung Vascular Endothelial Dysfunction Caused by Truncated Oxidized Phospholipids via Antagonism with CD36 Receptor. Am J Respir Cell Mol Biol 2024; 70:11-25. [PMID: 37725486 PMCID: PMC10768836 DOI: 10.1165/rcmb.2023-0127oc] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 09/12/2023] [Indexed: 09/21/2023] Open
Abstract
The generation of bioactive truncated oxidized phospholipids (Tr-OxPLs) from oxidation of cell-membrane or circulating lipoproteins is a common feature of various pathological states. Scavenger receptor CD36 is involved in lipid transport and acts as a receptor for Tr-OxPLs. Interestingly, Tr-OxPLs and CD36 are involved in endothelial dysfunction-derived acute lung injury, but the precise mechanistic connections remain unexplored. In the present study, we investigated the role of CD36 in mediating pulmonary endothelial cell (EC) dysfunction caused by Tr-OxPLs. Our results demonstrated that the Tr-OxPLs KOdia-PC, Paz-PC, PGPC, PON-PC, POV-PC, and lysophosphocholine caused an acute EC barrier disruption as revealed by measurements of transendothelial electrical resistance and VE-cadherin immunostaining. More importantly, a synthetic amphipathic helical peptide, L37pA, targeting human CD36 strongly attenuated Tr-OxPL-induced EC permeability. L37pA also suppressed Tr-OxPL-induced endothelial inflammatory activation monitored by mRNA expression of inflammatory cytokines/chemokines and adhesion molecules. In addition, L37pA blocked Tr-OxPL-induced NF-κB activation and tyrosine phosphorylation of Src kinase and VE-cadherin. The Src inhibitor SU6656 attenuated KOdia-PC-induced EC permeability and inflammation, but inhibition of the Toll-like receptors (TLRs) TLR1, TLR2, TLR4, and TLR6 had no such protective effects. CD36-knockout mice were more resistant to Tr-OxPL-induced lung injury. Treatment with L37pA was equally effective in ameliorating Tr-OxPL-induced vascular leak and lung inflammation as determined by an Evans blue extravasation assay and total cell and protein content in BAL fluid. Altogether, these results demonstrate an essential role of CD36 in mediating Tr-OxPL-induced EC dysfunction and suggest a strong therapeutic potential of CD36 inhibitory peptides in mitigating lung injury and inflammation.
Collapse
Affiliation(s)
- Pratap Karki
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Yue Li
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Chen-Ou Zhang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Yunbo Ke
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Kamoltip Promnares
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Anna A. Birukova
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Thomas L. Eggerman
- Department of Laboratory Medicine, Clinical Center, and
- National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | | | - Konstantin G. Birukov
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
10
|
Breslin JW. Edema and lymphatic clearance: molecular mechanisms and ongoing challenges. Clin Sci (Lond) 2023; 137:1451-1476. [PMID: 37732545 PMCID: PMC11025659 DOI: 10.1042/cs20220314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 08/18/2023] [Accepted: 08/31/2023] [Indexed: 09/22/2023]
Abstract
Resolution of edema remains a significant clinical challenge. Conditions such as traumatic shock, sepsis, or diabetes often involve microvascular hyperpermeability, which leads to tissue and organ dysfunction. Lymphatic insufficiency due to genetic causes, surgical removal of lymph nodes, or infections, leads to varying degrees of tissue swelling that impair mobility and immune defenses. Treatment options are limited to management of edema as there are no specific therapeutics that have demonstrated significant success for ameliorating microvascular leakage or impaired lymphatic function. This review examines current knowledge about the physiological, cellular, and molecular mechanisms that control microvascular permeability and lymphatic clearance, the respective processes for interstitial fluid formation and removal. Clinical conditions featuring edema, along with potential future directions are discussed.
Collapse
Affiliation(s)
- Jerome W Breslin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, FL, U.S.A
| |
Collapse
|
11
|
Wang C, Du M, Jiang Z, Cong R, Wang W, Zhang G, Li L. Comparative proteomic and phosphoproteomic analysis reveals differential heat response mechanism in two congeneric oyster species. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 263:115197. [PMID: 37451098 DOI: 10.1016/j.ecoenv.2023.115197] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 06/17/2023] [Accepted: 06/26/2023] [Indexed: 07/18/2023]
Abstract
High-temperature stress caused by global climate change poses a significant threat to marine ectotherms. This study investigated the role of protein phosphorylation modifications in the molecular regulation network under heat stress in oysters, which are representative intertidal organisms that experience considerable temperature changes. Firstly, the study compared the extent of thermal damage between two congeneric oyster species, the relative heat-tolerant Crassostrea angulata (C. angulata) and heat-sensitive Crassostrea gigas (C. gigas), under sublethal temperature (37 °C) for 12 h, using various physiological and biochemical methods. Subsequently, the comparative proteomic and phosphoproteomic analyses revealed that high-temperature considerably regulated signal transduction, energy metabolism, protein synthesis, cell survival and apoptosis, and cytoskeleton remodeling through phosphorylation modifications of related receptors and kinases. Furthermore, the protein kinase A, mitogen-activated protein kinase 1, tyrosine-protein kinase Src, and serine/threonine kinase AKT, exhibiting differential phosphorylation modification patterns, were identified as hub regulators that may enhance glycolysis and TCA cycle to increase the energy supply, distribute protein synthesis, inhibit Caspase-dependent apoptosis activated by endogenous mitochondrial cytochrome release and maintain cytoskeletal stability, ultimately shaping the higher thermal resistance of C. angulata. This study represents the first investigation of protein phosphorylation dynamics in marine invertebrates under heat stress, reveals the molecular mechanisms underlying the differential thermal responses between two Crassostrea oysters at the phosphorylation level, and provides new insights into understanding phosphorylation-mediated molecular responses in marine organisms during environmental changes and predicting the adaptive potential in the context of global warming.
Collapse
Affiliation(s)
- Chaogang Wang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Laoshan Laboratory, Qingdao, China; University of Chinese Academy of Sciences, Beijing, China
| | - Mingyang Du
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Laoshan Laboratory, Qingdao, China; University of Chinese Academy of Sciences, Beijing, China
| | - Zhuxiang Jiang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Laoshan Laboratory, Qingdao, China; University of Chinese Academy of Sciences, Beijing, China
| | - Rihao Cong
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Laoshan Laboratory, Qingdao, China; National and Local Joint Engineering Laboratory of Ecological Mariculture, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; Shandong Technology Innovation Center of Oyster Seed Industry, Qingdao, China
| | - Wei Wang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Fisheries Science and Food Production Processes, Laoshan Laboratory, Qingdao, China; National and Local Joint Engineering Laboratory of Ecological Mariculture, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; Shandong Technology Innovation Center of Oyster Seed Industry, Qingdao, China
| | - Guofan Zhang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Laoshan Laboratory, Qingdao, China; Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; National and Local Joint Engineering Laboratory of Ecological Mariculture, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; Shandong Technology Innovation Center of Oyster Seed Industry, Qingdao, China
| | - Li Li
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; University of Chinese Academy of Sciences, Beijing, China; Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; Laboratory for Marine Fisheries Science and Food Production Processes, Laoshan Laboratory, Qingdao, China; National and Local Joint Engineering Laboratory of Ecological Mariculture, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; Shandong Technology Innovation Center of Oyster Seed Industry, Qingdao, China.
| |
Collapse
|
12
|
Baoyinna B, Miao J, Oliver PJ, Ye Q, Shaheen N, Kalin T, He J, Parinandi NL, Zhao Y, Zhao J. Non-Lethal Doses of RSL3 Impair Microvascular Endothelial Barrier through Degradation of Sphingosie-1-Phosphate Receptor 1 and Cytoskeletal Arrangement in A Ferroptosis-Independent Manner. Biomedicines 2023; 11:2451. [PMID: 37760892 PMCID: PMC10525432 DOI: 10.3390/biomedicines11092451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 08/28/2023] [Accepted: 09/02/2023] [Indexed: 09/29/2023] Open
Abstract
The excess microvascular endothelial permeability is a hallmark of acute inflammatory diseases. Maintenance of microvascular integrity is critical to preventing leakage of vascular components into the surrounding tissues. Sphingosine-1-phosphate (S1P) is an active lysophospholipid that enhances the endothelial cell (EC) barrier via activation of its receptor S1PR1. Here, we delineate the effect of non-lethal doses of RSL3, an inhibitor of glutathione peroxidase 4 (GPX4), on EC barrier function. Low doses of RSL3 (50-100 nM) attenuated S1P-induced human lung microvascular barrier enhancement and the phosphorylation of AKT. To investigate the molecular mechanisms by which RSL3 attenuates S1P's effect, we examined the S1PR1 levels. RSL3 treatment reduced S1PR1 levels in 1 h, whereas the effect was attenuated by the proteasome and lysosome inhibitors as well as a lipid raft inhibitor. Immunofluorescence staining showed that RSL3 induced S1PR1 internalization from the plasma membrane into the cytoplasm. Furthermore, we found that RSL3 (100 and 200 nM) increased EC barrier permeability and cytoskeletal rearrangement without altering cell viability. Taken together, our data delineates that non-lethal doses of RSL3 impair EC barrier function via two mechanisms. RSL3 attenuates S1P1-induced EC barrier enhancement and disrupts EC barrier integrity through the generation of 4-hydroxynonena (4HNE). All these effects are independent of ferroptosis.
Collapse
Affiliation(s)
- Boina Baoyinna
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Jiaxing Miao
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Patrick J. Oliver
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Qinmao Ye
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Nargis Shaheen
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Timothy Kalin
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Jinshan He
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | | | - Yutong Zhao
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Jing Zhao
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
13
|
Kushwaha R, Li Y, Makarava N, Pandit NP, Molesworth K, Birukov KG, Baskakov IV. Reactive astrocytes associated with prion disease impair the blood brain barrier. Neurobiol Dis 2023; 185:106264. [PMID: 37597815 PMCID: PMC10494928 DOI: 10.1016/j.nbd.2023.106264] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 07/31/2023] [Accepted: 08/15/2023] [Indexed: 08/21/2023] Open
Abstract
BACKGROUND Impairment of the blood-brain barrier (BBB) is considered to be a common feature among neurodegenerative diseases, including Alzheimer's, Parkinson's and prion diseases. In prion disease, increased BBB permeability was reported 40 years ago, yet the mechanisms behind the loss of BBB integrity have never been explored. Recently, we showed that reactive astrocytes associated with prion diseases are neurotoxic. The current work examines the potential link between astrocyte reactivity and BBB breakdown. RESULTS In prion-infected mice, the loss of BBB integrity and aberrant localization of aquaporin 4 (AQP4), a sign of retraction of astrocytic endfeet from blood vessels, were noticeable prior to disease onset. Gaps in cell-to-cell junctions along blood vessels, together with downregulation of Occludin, Claudin-5 and VE-cadherin, which constitute tight and adherens junctions, suggested that loss of BBB integrity is linked with degeneration of vascular endothelial cells. In contrast to cells isolated from non-infected adult mice, endothelial cells originating from prion-infected mice displayed disease-associated changes, including lower levels of Occludin, Claudin-5 and VE-cadherin expression, impaired tight and adherens junctions, and reduced trans-endothelial electrical resistance (TEER). Endothelial cells isolated from non-infected mice, when co-cultured with reactive astrocytes isolated from prion-infected animals or treated with media conditioned by the reactive astrocytes, developed the disease-associated phenotype observed in the endothelial cells from prion-infected mice. Reactive astrocytes were found to produce high levels of secreted IL-6, and treatment of endothelial monolayers originating from non-infected animals with recombinant IL-6 alone reduced their TEER. Remarkably, treatment with extracellular vesicles produced by normal astrocytes partially reversed the disease phenotype of endothelial cells isolated from prion-infected animals. CONCLUSIONS To our knowledge, the current work is the first to illustrate early BBB breakdown in prion disease and to document that reactive astrocytes associated with prion disease are detrimental to BBB integrity. Moreover, our findings suggest that the harmful effects are linked to proinflammatory factors secreted by reactive astrocytes.
Collapse
Affiliation(s)
- Rajesh Kushwaha
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201, United States of America; Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, United States of America
| | - Yue Li
- Lung Biology Research Program and Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD 21201, United States of America
| | - Natallia Makarava
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201, United States of America; Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, United States of America
| | - Narayan P Pandit
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201, United States of America; Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, United States of America
| | - Kara Molesworth
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201, United States of America; Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, United States of America
| | - Konstantin G Birukov
- Lung Biology Research Program and Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD 21201, United States of America
| | - Ilia V Baskakov
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201, United States of America; Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, United States of America.
| |
Collapse
|
14
|
Karki P, Zhang CO, Promnares K, Li Y, Ke Y, Birukova AA, Birukov KG. Truncated oxidized phospholipids exacerbate endothelial dysfunction and lung injury caused by bacterial pathogens. Cell Signal 2023; 109:110804. [PMID: 37437826 PMCID: PMC10544726 DOI: 10.1016/j.cellsig.2023.110804] [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: 04/21/2023] [Revised: 07/06/2023] [Accepted: 07/08/2023] [Indexed: 07/14/2023]
Abstract
Oxidized phospholipids (OxPLs) are present at basal levels in circulation of healthy individuals, but a substantial increase and changes in composition of OxPLs may rapidly occur during microbial infections, sepsis, and trauma. Specifically, truncated oxidized phospholipids (Tr-OxPLs) exhibit detrimental effects on pulmonary endothelium, yet their role on modulation of lung injury caused by bacterial pathogens remains to be elucidated. This study investigated the effects of Tr-OxPL species: KOdiA-PC, POV-PC, PON-PC, PAz-PC, PGPC, and Lyso-PC on endothelial permeability and inflammatory responses to gram-positive bacterial particles. Results showed that all six tested Tr-OxPLs augmented endothelial barrier disruption caused by heat-killed Staphylococcus aureus (HKSA) as determined by VE-cadherin immunostaining and monitoring transendothelial electrical resistance. In parallel, even moderate elevation of Tr-OxPLs augmented HKSA-induced activation of NF-κB, secretion of IL-6 and IL-8, and protein expression of ICAM-1 and VCAM-1. In the mouse model of acute lung injury caused by intranasal injection of HKSA, intravenous Tr-OxPLs administration augmented HKSA-induced increase in BAL protein content and cell counts, tissue expression of TNFα, KC, IL1β, and CCL2, and promoted vascular leak monitored by lung infiltration of Evans Blue. These results suggest that elevated Tr-OxPLs act as critical risk factor worsening bacterial pathogen-induced endothelial dysfunction and lung injury.
Collapse
Affiliation(s)
- Pratap Karki
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, United States of America.
| | - Chen-Ou Zhang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, United States of America
| | - Kamoltip Promnares
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD 21201, United States of America
| | - Yue Li
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, United States of America
| | - Yunbo Ke
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD 21201, United States of America
| | - Anna A Birukova
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, United States of America
| | - Konstantin G Birukov
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD 21201, United States of America
| |
Collapse
|
15
|
Khapchaev AY, Antonova OA, Kazakova OA, Samsonov MV, Vorotnikov AV, Shirinsky VP. Long-Term Experimental Hyperglycemia Does Not Impair Macrovascular Endothelial Barrier Integrity and Function in vitro. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:1126-1138. [PMID: 37758312 DOI: 10.1134/s0006297923080072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 07/08/2023] [Accepted: 07/11/2023] [Indexed: 10/03/2023]
Abstract
Hyperglycemia is a hallmark of type 2 diabetes implicated in vascular endothelial dysfunction and cardiovascular complications. Many in vitro studies identified endothelial apoptosis as an early outcome of experimentally modeled hyperglycemia emphasizing cell demise as a significant factor of vascular injury. However, endothelial apoptosis has not been observed in vivo until the late stages of type 2 diabetes. Here, we studied the long-term (up to 4 weeks) effects of high glucose (HG, 30 mM) on human umbilical vein endothelial cells (HUVEC) in vitro. HG did not alter HUVEC monolayer morphology, ROS levels, NO production, and exerted minor effects on the HUVEC apoptosis markers. The barrier responses to various clues were indistinguishable from those by cells cultured in physiological glucose (5 mM). Tackling the key regulators of cytoskeletal contractility and endothelial barrier revealed no differences in the histamine-induced intracellular Ca2+ responses, nor in phosphorylation of myosin regulatory light chain or myosin light chain phosphatase. Altogether, these findings suggest that vascular endothelial cells may well tolerate HG for relatively long exposures and warrant further studies to explore mechanisms involved in vascular damage in advanced type 2 diabetes.
Collapse
Affiliation(s)
- Asker Y Khapchaev
- Institute of Experimental Cardiology, Chazov National Medical Research Center for Cardiology, Moscow, 121552, Russia.
| | - Olga A Antonova
- Institute of Experimental Cardiology, Chazov National Medical Research Center for Cardiology, Moscow, 121552, Russia
| | - Olga A Kazakova
- Institute of Experimental Cardiology, Chazov National Medical Research Center for Cardiology, Moscow, 121552, Russia
| | - Mikhail V Samsonov
- Institute of Experimental Cardiology, Chazov National Medical Research Center for Cardiology, Moscow, 121552, Russia
| | - Alexander V Vorotnikov
- Institute of Experimental Cardiology, Chazov National Medical Research Center for Cardiology, Moscow, 121552, Russia
| | - Vladimir P Shirinsky
- Institute of Experimental Cardiology, Chazov National Medical Research Center for Cardiology, Moscow, 121552, Russia
| |
Collapse
|
16
|
Kleinjan ML, Mao DY, Naiche LA, Joshi JC, Gupta A, Jesse JJ, Shaye DD, Mehta D, Kitajewski J. CLIC4 Regulates Endothelial Barrier Control by Mediating PAR1 Signaling via RhoA. Arterioscler Thromb Vasc Biol 2023; 43:1441-1454. [PMID: 37317855 PMCID: PMC10527476 DOI: 10.1161/atvbaha.123.319206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 05/30/2023] [Indexed: 06/16/2023]
Abstract
BACKGROUND Endothelial CLICs (chloride intracellular channel proteins) CLIC1 and CLIC4 are required for the GPCRs (G-protein-coupled receptors) S1PR1 (sphingosine-1-phosphate receptor 1) and S1PR3 to activate the small GTPases Rac1 (Ras-related C3 botulinum toxin substrate 1) and RhoA (Ras homolog family member A). To determine whether CLIC1 and CLIC4 function in additional endothelial GPCR pathways, we evaluated CLIC function in thrombin signaling via the thrombin-regulated PAR1 (protease-activated receptor 1) and downstream effector RhoA. METHODS We assessed the ability of CLIC1 and CLIC4 to relocalize to cell membranes in response to thrombin in human umbilical vein endothelial cells (HUVEC). We examined CLIC1 and CLIC4 function in HUVEC by knocking down expression of each CLIC protein and compared thrombin-mediated RhoA or Rac1 activation, ERM (ezrin/radixin/moesin) phosphorylation, and endothelial barrier modulation in control and CLIC knockdown HUVEC. We generated a conditional murine allele of Clic4 and examined PAR1-mediated lung microvascular permeability and retinal angiogenesis in mice with endothelial-specific loss of Clic4. RESULTS Thrombin promoted relocalization of CLIC4, but not CLIC1, to HUVEC membranes. Knockdown of CLIC4 in HUVEC reduced thrombin-mediated RhoA activation, ERM phosphorylation, and endothelial barrier disruption. Knockdown of CLIC1 did not reduce thrombin-mediated RhoA activity but prolonged the RhoA and endothelial barrier response to thrombin. Endothelial-specific deletion of Clic4 in mice reduced lung edema and microvascular permeability induced by PAR1 activating peptide. CONCLUSIONS CLIC4 is a critical effector of endothelial PAR1 signaling and is required to regulate RhoA-mediated endothelial barrier disruption in cultured endothelial cells and murine lung endothelium. CLIC1 was not critical for thrombin-mediated barrier disruption but contributed to the barrier recovery phase after thrombin treatment.
Collapse
Affiliation(s)
- Matthew L. Kleinjan
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| | - De Yu Mao
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| | - L. A. Naiche
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| | - Jagdish C. Joshi
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL, USA
| | - Ahana Gupta
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| | - Jordan J. Jesse
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| | - Daniel D. Shaye
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| | - Dolly Mehta
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL, USA
| | - Jan Kitajewski
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA
- University of Illinois Cancer Center, Chicago, IL, USA
| |
Collapse
|
17
|
Ke Y, Karki P, Li Y, Promnares K, Zhang CO, Eggerman TL, Bocharov AV, Birukova AA, Birukov KG. Aging-Related Accumulation of Truncated Oxidized Phospholipids Augments Infectious Lung Injury and Endothelial Dysfunction via Cluster of Differentiation 36-Dependent Mechanism. Cells 2023; 12:1937. [PMID: 37566016 PMCID: PMC10416939 DOI: 10.3390/cells12151937] [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: 04/04/2023] [Revised: 07/18/2023] [Accepted: 07/24/2023] [Indexed: 08/12/2023] Open
Abstract
Truncated phospholipid oxidation products (Tr-OxPL) increase in blood circulation with aging; however, their role in the severity of vascular dysfunction and bacterial lung injury in aging groups remains poorly understood. We investigated the effects of six Tr-OxPL species: KOdiA-PC, POVPC, PONPC, PGPC, Paz-PC, and Lyso-PC on endothelial dysfunction and lung inflammation caused by heat-killed Staphylococcus aureus (HKSA) in young (aged 2-4 months) and old (aged 12-18 months) mice, organotypic culture of precisely cut lung slices, and endothelial cells (mLEC) isolated from young and old mice. HKSA and Tr-OxPL combination caused a higher degree of vascular leak, the accumulation of inflammatory cells and protein in bronchoalveolar lavage, and inflammatory gene expression in old mice lungs. HKSA caused a greater magnitude of inflammatory gene activation in cell and ex vivo cultures from old mice, which was further augmented by Tr-OxPLs. L37pA peptide targeting CD36 receptor attenuated Tr-OxPL-induced endothelial cell permeability in young and old mLEC and ameliorated KOdiA-PC-induced vascular leak and lung inflammation in vivo. Finally, CD36 knockout mice showed better resistance to KOdiA-PC-induced lung injury in both age groups. These results demonstrate the aging-dependent vulnerability of pulmonary vasculature to elevated Tr-OxPL, which exacerbates bacterial lung injury. CD36 inhibition is a promising therapeutic approach for improving pneumonia outcomes in aging population.
Collapse
Affiliation(s)
- Yunbo Ke
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Pratap Karki
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (P.K.)
| | - Yue Li
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (P.K.)
| | - Kamoltip Promnares
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Chen-Ou Zhang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (P.K.)
| | - Thomas L. Eggerman
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20894, USA
- National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Alexander V. Bocharov
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20894, USA
| | - Anna A. Birukova
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (P.K.)
| | - Konstantin G. Birukov
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
18
|
Patil RS, Kovacs-Kasa A, Gorshkov BA, Fulton DJR, Su Y, Batori RK, Verin AD. Serine/Threonine Protein Phosphatases 1 and 2A in Lung Endothelial Barrier Regulation. Biomedicines 2023; 11:1638. [PMID: 37371733 PMCID: PMC10296329 DOI: 10.3390/biomedicines11061638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 05/28/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
Vascular barrier dysfunction is characterized by increased permeability and inflammation of endothelial cells (ECs), which are prominent features of acute lung injury (ALI), acute respiratory distress syndrome (ARDS), and sepsis, and a major complication of the SARS-CoV-2 infection and COVID-19. Functional impairment of the EC barrier and accompanying inflammation arises due to microbial toxins and from white blood cells of the lung as part of a defensive action against pathogens, ischemia-reperfusion or blood product transfusions, and aspiration syndromes-based injury. A loss of barrier function results in the excessive movement of fluid and macromolecules from the vasculature into the interstitium and alveolae resulting in pulmonary edema and collapse of the architecture and function of the lungs, and eventually culminates in respiratory failure. Therefore, EC barrier integrity, which is heavily dependent on cytoskeletal elements (mainly actin filaments, microtubules (MTs), cell-matrix focal adhesions, and intercellular junctions) to maintain cellular contacts, is a critical requirement for the preservation of lung function. EC cytoskeletal remodeling is regulated, at least in part, by Ser/Thr phosphorylation/dephosphorylation of key cytoskeletal proteins. While a large body of literature describes the role of phosphorylation of cytoskeletal proteins on Ser/Thr residues in the context of EC barrier regulation, the role of Ser/Thr dephosphorylation catalyzed by Ser/Thr protein phosphatases (PPases) in EC barrier regulation is less documented. Ser/Thr PPases have been proposed to act as a counter-regulatory mechanism that preserves the EC barrier and opposes EC contraction. Despite the importance of PPases, our knowledge of the catalytic and regulatory subunits involved, as well as their cellular targets, is limited and under-appreciated. Therefore, the goal of this review is to discuss the role of Ser/Thr PPases in the regulation of lung EC cytoskeleton and permeability with special emphasis on the role of protein phosphatase 1 (PP1) and protein phosphatase 2A (PP2A) as major mammalian Ser/Thr PPases. Importantly, we integrate the role of PPases with the structural dynamics of the cytoskeleton and signaling cascades that regulate endothelial cell permeability and inflammation.
Collapse
Affiliation(s)
- Rahul S. Patil
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Anita Kovacs-Kasa
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Boris A. Gorshkov
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - David J. R. Fulton
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Department of Pharmacology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Yunchao Su
- Department of Pharmacology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Robert K. Batori
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Alexander D. Verin
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
19
|
Kushwaha R, Li Y, Makarava N, Pandit NP, Molesworth K, Birukov KG, Baskakov IV. Reactive astrocytes associated with prion disease impair the blood brain barrier. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.21.533684. [PMID: 36993690 PMCID: PMC10055297 DOI: 10.1101/2023.03.21.533684] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Background Impairment of the blood-brain barrier (BBB) is considered to be a common feature among neurodegenerative diseases, including Alzheimer's, Parkinson's and prion diseases. In prion disease, increased BBB permeability was reported 40 years ago, yet the mechanisms behind the loss of BBB integrity have never been explored. Recently, we showed that reactive astrocytes associated with prion diseases are neurotoxic. The current work examines the potential link between astrocyte reactivity and BBB breakdown. Results In prion-infected mice, the loss of BBB integrity and aberrant localization of aquaporin 4 (AQP4), a sign of retraction of astrocytic endfeet from blood vessels, were noticeable prior to disease onset. Gaps in cell-to-cell junctions along blood vessels, together with downregulation of Occludin, Claudin-5 and VE-cadherin, which constitute tight and adherens junctions, suggested that loss of BBB integrity is linked with degeneration of vascular endothelial cells. In contrast to cells isolated from non-infected adult mice, endothelial cells originating from prion-infected mice displayed disease-associated changes, including lower levels of Occludin, Claudin-5 and VE-cadherin expression, impaired tight and adherens junctions, and reduced trans-endothelial electrical resistance (TEER). Endothelial cells isolated from non-infected mice, when co-cultured with reactive astrocytes isolated from prion-infected animals or treated with media conditioned by the reactive astrocytes, developed the disease-associated phenotype observed in the endothelial cells from prion-infected mice. Reactive astrocytes were found to produce high levels of secreted IL-6, and treatment of endothelial monolayers originating from non-infected animals with recombinant IL-6 alone reduced their TEER. Remarkably, treatment with extracellular vesicles produced by normal astrocytes partially reversed the disease phenotype of endothelial cells isolated from prion-infected animals. Conclusions To our knowledge, the current work is the first to illustrate early BBB breakdown in prion disease and to document that reactive astrocytes associated with prion disease are detrimental to BBB integrity. Moreover, our findings suggest that the harmful effects are linked to proinflammatory factors secreted by reactive astrocytes.
Collapse
Affiliation(s)
- Rajesh Kushwaha
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, 21201, United States of America
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, 21201, United States of America
| | - Yue Li
- Lung Biology Research Program and Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, 21201
| | - Natallia Makarava
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, 21201, United States of America
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, 21201, United States of America
| | - Narayan P. Pandit
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, 21201, United States of America
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, 21201, United States of America
| | - Kara Molesworth
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, 21201, United States of America
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, 21201, United States of America
| | - Konstantin G. Birukov
- Lung Biology Research Program and Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, 21201
| | - Ilia V. Baskakov
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, 21201, United States of America
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, 21201, United States of America
| |
Collapse
|
20
|
Deng Y, Huang X, Hu Y, Zhong W, Zhang H, Mo C, Wang H, Ding BS, Wang C. Deficiency of endothelial FGFR1 signaling via upregulation of ROCK2 activity aggravated ALI/ARDS. Front Immunol 2023; 14:1041533. [PMID: 36969192 PMCID: PMC10036754 DOI: 10.3389/fimmu.2023.1041533] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 02/13/2023] [Indexed: 03/12/2023] Open
Abstract
Vascular leakage and inflammation are pathological hallmarks of acute lung injury (ALI)/acute respiratory distress syndrome (ARDS). Endothelial cells (ECs) serve as a semipermeable barrier and play a key role in disease progression. It is well known that fibroblast growth factor receptor 1 (FGFR1) is required for maintaining vascular integrity. However, how endothelial FGFR1 functions in ALI/ARDS remains obscure. Here, we revealed that conditional deletion of endothelial FGFR1 aggravated LPS-induced lung injury, including inflammation and vascular leakage. Inhibition of its downstream Rho-associated coiled-coil–forming protein kinase 2 (ROCK2) by AAV Vec-tie-shROCK2 or its selective inhibitor TDI01 effectively attenuated inflammation and vascular leakage in a mouse model. In vitro, TNFα-stimulated human umbilical vein endothelial cells (HUVECs) showed decreased FGFR1 expression and increased ROCK2 activity. Furthermore, knockdown of FGFR1 activated ROCK2 and thus promoted higher adhesive properties to inflammatory cells and higher permeability in HUVECs. TDI01 effectively suppressed ROCK2 activity and rescued the endothelial dysfunction. These data demonstrated that the loss of endothelial FGFR1 signaling mediated an increase in ROCK2 activity, which led to an inflammatory response and vascular leakage in vivo and in vitro. Moreover, inhibition of ROCK2 activity by TDI01 provided great value and shed light on clinical translation.
Collapse
Affiliation(s)
- Yue Deng
- Peking University China–Japan Friendship School of Clinical Medicine, Beijing, China
- National Center for Respiratory Medicine, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, National Clinical Research Center for Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China
| | - Xingming Huang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yan Hu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Weiting Zhong
- Beijing Tide Pharmaceutical Co., Ltd., Beijing, China
| | - Hua Zhang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Chunheng Mo
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Hongjun Wang
- Beijing Tide Pharmaceutical Co., Ltd., Beijing, China
- *Correspondence: Chen Wang, ; Bi-Sen Ding, ; Hongjun Wang,
| | - Bi-Sen Ding
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
- *Correspondence: Chen Wang, ; Bi-Sen Ding, ; Hongjun Wang,
| | - Chen Wang
- Peking University China–Japan Friendship School of Clinical Medicine, Beijing, China
- National Center for Respiratory Medicine, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, National Clinical Research Center for Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China
- State Key Laboratory of Medical Molecular Biology, Department of Physiology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
- *Correspondence: Chen Wang, ; Bi-Sen Ding, ; Hongjun Wang,
| |
Collapse
|
21
|
Xie Q, Liu R, Zou Z, Feng Y, Huang Y, Xu G, Sun W, Liang Y, Zhong W. MYPT1 inhibits the metastasis of renal clear cell carcinoma via the MAPK8/N-cadherin pathway. FEBS Open Bio 2022; 12:2083-2095. [PMID: 36106411 PMCID: PMC9623519 DOI: 10.1002/2211-5463.13487] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 08/23/2022] [Accepted: 09/14/2022] [Indexed: 01/25/2023] Open
Abstract
Myosin phosphatase target subunit 1 (MYPT1) is a subunit of myosin phosphatase that is capable of regulating smooth muscle contraction. MYPT1 has been reported to be involved in a wide variety of tumours, but its expression and biological functions in renal clear cell carcinoma (ccRCC) remain obscure. Herein, we analysed the relationship between patient clinicopathological characteristics and MYPT1 expression levels in ccRCC patients using a tissue microarray (TMA) and data retrieved from the TCGA-KIRC dataset. MYPT1 was overexpressed or depleted using siRNA in ccRCC cells to assess the effects on migration and invasion in vitro and in vivo. Additionally, RNA-sequencing and bioinformatics analysis were performed to investigate the precise mechanism. MYPT1 expression in ccRCC tissues was observed to be lower than that in nonmalignant tissues (P < 0.05). In addition, MYPT1 downregulation was closely linked to advanced pathological stage (P < 0.05), and poor OS (overall survival; P < 0.05). Functionally, increased expression of MYPT1 suppressed ccRCC migration and invasion in vitro, and inhibited tumour metastasis in vivo. In addition, MYPT1 overexpression exerted its suppressive effects via the MAPK8/N-cadherin pathway in ccRCC.
Collapse
Affiliation(s)
- Qingling Xie
- Guangdong Provincial Institute of Nephrology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Ren Liu
- Guangdong Provincial Institute of Nephrology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Zhihao Zou
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of MedicineSouth China University of TechnologyGuangzhouChina
| | - Yuanfa Feng
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of MedicineSouth China University of TechnologyGuangzhouChina
| | - Yiqiao Huang
- Department of UrologyThe Fifth Affiliated Hospital of Guangzhou Medical UniversityChina
| | - Guibin Xu
- Department of UrologyThe Fifth Affiliated Hospital of Guangzhou Medical UniversityChina
| | - Wei Sun
- Department of Urology, Huadu District People's HospitalSouthern Medical UniversityGuangzhouChina
| | - Yuxiang Liang
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of MedicineSouth China University of TechnologyGuangzhouChina
| | - Weide Zhong
- Guangdong Provincial Institute of Nephrology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina,Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of MedicineSouth China University of TechnologyGuangzhouChina
| |
Collapse
|
22
|
Mauerhofer C, Afonyushkin T, Oskolkova OV, Hellauer K, Gesslbauer B, Schmerda J, Ke Y, Zimmer A, Birukova AA, Birukov KG, Bochkov V. Low Concentrations of Oxidized Phospholipids Increase Stress Tolerance of Endothelial Cells. Antioxidants (Basel) 2022; 11:1741. [PMID: 36139816 PMCID: PMC9495896 DOI: 10.3390/antiox11091741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/19/2022] [Accepted: 08/24/2022] [Indexed: 12/15/2022] Open
Abstract
Oxidized phospholipids (OxPLs) are generated by enzymatic or autooxidation of esterified polyunsaturated fatty acids (PUFAs) residues. OxPLs are present in circulation and atherosclerotic plaques where they are thought to induce predominantly proinflammatory and toxic changes in endothelial (ECs) and other cell types. Unexpectedly, we found that low concentrations of OxPLs were not toxic but protected ECs from stress induced by serum deprivation or cytostatic drugs. The protective effect was observed in ECs obtained from different vessels and was monitored using a variety of readouts based on different biological and chemical principles. Analysis of the structure−activity relationship identified oxidized or missing fatty acid residue (OxPLs or Lyso-PLs, respectively) as a prerequisite for the protective action of a PL. Protective OxPLs or Lyso-PLs acquired detergent-like properties and formed in solution aggregates <10 nm in diameter (likely micelles), which were in striking contrast with large aggregates (>1000 nm, likely multilayer liposomes) produced by nonoxidized precursor PLs. Because surfactants, OxPLs, and Lyso-PLs are known to extract membrane cholesterol, we tested if this effect might trigger the protection of endothelial cells. The protective action of OxPLs and Lyso-PLs was inhibited by cotreatment with cholesterol and mimicked by cholesterol-binding beta-cyclodextrin but not inactive α-cyclodextrin. Wide-scale mRNA expression analysis in four types of ECs showed the induction of genes encoding for heat shock proteins (HSPs) and secreted prosurvival peptides and proteins. Inducers of HSPs, chemical chaperones, and pure prosurvival factors mimicked the protective action of OxPLs/Lyso-PLs. We hypothesize that oxidation changes the physicochemical properties of PLs, thus promoting membrane cholesterol redistribution or extraction leading to the expression of intra- and extracellular prosurvival factors.
Collapse
Affiliation(s)
- Christina Mauerhofer
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Graz, Humboldtstrasse 46/III, 8010 Graz, Austria
| | - Taras Afonyushkin
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse 17, 1090 Vienna, Austria
| | - Olga V. Oskolkova
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Graz, Humboldtstrasse 46/III, 8010 Graz, Austria
| | - Klara Hellauer
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Graz, Humboldtstrasse 46/III, 8010 Graz, Austria
| | - Bernd Gesslbauer
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Graz, Humboldtstrasse 46/III, 8010 Graz, Austria
| | - Jasmin Schmerda
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Graz, Humboldtstrasse 46/III, 8010 Graz, Austria
| | - Yunbo Ke
- Department of Anesthesiology, University of Maryland School of Medicine, 20 Penn. Street, HSF-2, Room 145, Baltimore, MD 21201, USA
| | - Andreas Zimmer
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical Technology and Biopharmacy, University of Graz, Universitätsplatz 1/EG, 8010 Graz, Austria
| | - Anna A. Birukova
- Department of Anesthesiology, University of Maryland School of Medicine, 20 Penn. Street, HSF-2, Room 145, Baltimore, MD 21201, USA
| | - Konstantin G. Birukov
- Department of Anesthesiology, University of Maryland School of Medicine, 20 Penn. Street, HSF-2, Room 145, Baltimore, MD 21201, USA
| | - Valery Bochkov
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Graz, Humboldtstrasse 46/III, 8010 Graz, Austria
- Field of Excellence BioHealth, University of Graz, 8010 Graz, Austria
| |
Collapse
|
23
|
Kim J, Baalachandran R, Li Y, Zhang CO, Ke Y, Karki P, Birukov KG, Birukova AA. Circulating extracellular histones exacerbate acute lung injury by augmenting pulmonary endothelial dysfunction via TLR4-dependent mechanism. Am J Physiol Lung Cell Mol Physiol 2022; 323:L223-L239. [PMID: 35852995 PMCID: PMC9512107 DOI: 10.1152/ajplung.00072.2022] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 06/21/2022] [Accepted: 06/27/2022] [Indexed: 11/22/2022] Open
Abstract
Extracellular histones released into the circulation following trauma, sepsis, and ARDS may act as potent damage-associated molecular pattern signals leading to multiple organ failure. Endothelial cell (EC) dysfunction caused by extracellular histones has been demonstrated in vitro and in vivo; however, precise mechanistic details of histone-induced EC dysfunction and exacerbation of ongoing inflammation remain poorly understood. This study investigated the role of extracellular histones in exacerbating preexisting endothelial dysfunction and acute lung injury. Histone subunits H3 and H4, but not H1, H2A, or H2B, induced permeability in human pulmonary EC. H3 and H4 at concentrations above 30 µg/mL caused EC inflammation reflected by activation of the NF-κB pathway, transcriptional activation, and release of cytokines and chemokines including IL-6 and IL-8, and increased mRNA and protein expression of EC adhesion molecules VCAM-1 and ICAM-1. Pharmacological inhibitors targeting Toll-like receptor TLR4 but not TLR2/6, blocked histone-induced EC dysfunction. H3 and H4 also strongly augmented EC permeability and inflammation caused by Gram-negative and Gram-positive bacterial particles, endotoxin, and TNFα. Heparin blocked histone-induced augmentation of EC inflammation caused by endotoxin and TNFα. Injection of histone in mouse models of lung injury caused by bacterial wall lipopolysaccharide (LPS) and heat-killed Staphylococcus aureus (HKSA) augmented ALI parameters: increased protein content, cell count, and inflammatory cytokine secretion in bronchoalveolar lavage fluid. Important clinical significance of these findings is in the demonstration that even a modest increase in extracellular histone levels can act as a severe exacerbating factor in conjunction with other EC barrier disruptive or proinflammatory agents.
Collapse
Affiliation(s)
- Junghyun Kim
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Ramasubramanian Baalachandran
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Yue Li
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Chen-Ou Zhang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Yunbo Ke
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Pratap Karki
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Konstantin G Birukov
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Anna A Birukova
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
24
|
Ni N, Wang W, Sun Y, Sun X, Leong DT. Inducible endothelial leakiness in nanotherapeutic applications. Biomaterials 2022; 287:121640. [PMID: 35772348 DOI: 10.1016/j.biomaterials.2022.121640] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 05/23/2022] [Accepted: 06/14/2022] [Indexed: 11/02/2022]
Abstract
All intravenous delivered nanomedicine needs to escape from the blood vessel to exert their therapeutic efficacy at their designated site of action. Failure to do so increases the possibility of detrimental side effects and negates their therapeutic intent. Many powerful anticancer nanomedicine strategies rely solely on the tumor derived enhanced permeability and retention (EPR) effect for the only mode of escaping from the tumor vasculature. However, not all tumors have the EPR effect nor can the EPR effect be induced or controlled for its location and timeliness. In recent years, there have been exciting developments along the lines of inducing endothelial leakiness at the tumor to decrease the dependence of EPR. Physical disruption of the endothelial-endothelial cell junctions with coordinated biological intrinsic pathways have been proposed that includes various modalities like ultrasound, radiotherapy, heat and even nanoparticles, appear to show good progress towards the goal of inducing endothelial leakiness. This review explains the intricate and complex biological background behind the endothelial cells with linkages on how updated reported nanomedicine strategies managed to induce endothelial leakiness. This review will also end off with fresh insights on where the future of inducible endothelial leakiness holds.
Collapse
Affiliation(s)
- Nengyi Ni
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| | - Weiyi Wang
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, China
| | - Yu Sun
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore; Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, No.88 Jiefang Road, Shangcheng District, Hangzhou, 310009, PR China
| | - Xiao Sun
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, China.
| | - David Tai Leong
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore.
| |
Collapse
|
25
|
Differential expression profile of plasma exosomal microRNAs in acute type A aortic dissection with acute lung injury. Sci Rep 2022; 12:11667. [PMID: 35804020 PMCID: PMC9270349 DOI: 10.1038/s41598-022-15859-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 06/30/2022] [Indexed: 11/08/2022] Open
Abstract
MicroRNAs (miRNAs) packaged into exosomes mediate cell communication and contribute to the pathogenesis of acute type A aortic dissection (ATAAD) with acute lung injury (ALI). The expression profile of plasma exosomal miRNAs in ATAAD patients with ALI hasn’t been identified. We performed a miRNA-sequencing to analyze the differentially expressed miRNAs (DE-miRNAs) of circulating exosomes in ATAAD patients with ALI compared to patients without ALI, founding 283 specific miRNAs in two groups. We respectively selected the top 10 downregulated and upregulated DE-miRNAs for further studies. The predicted transcription factors (TFs) of these DE-miRNAs were SMAD2, SRSF1, USF1, etc. The Gene Ontology (GO) and Kyoto Encyclopedia Genes and Genomes (KEGG) analysis predicted their target genes mainly involved acute inflammatory response, cell junction, cytoskeleton, NF-κB signaling pathway, etc. Construction and analysis of the PPI network revealed that RHOA and INSR were considered hub genes with the highest connectivity degrees. Moreover, we confirmed two exosomal miRNAs (hsa-miR-485-5p and hsa-miR-206) by real-time quantitative polymerase chain reaction (RT-qPCR) in a validation cohort. Our study identified a plasma exosomal miRNAs signature related to ATAAD with ALI. Certain DE-miRNAs may contribute to the progression of this disease, which help us better understand the pathogenesis of ATAAD with ALI.
Collapse
|
26
|
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]
|
27
|
Somatostatin Primes Endothelial Cells for Agonist-Induced Hyperpermeability and Angiogenesis In Vitro. Int J Mol Sci 2022; 23:ijms23063098. [PMID: 35328517 PMCID: PMC8949535 DOI: 10.3390/ijms23063098] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/08/2022] [Accepted: 03/09/2022] [Indexed: 02/04/2023] Open
Abstract
Somatostatin is an inhibitory peptide, which regulates the release of several hormones, and affects neurotransmission and cell proliferation via its five Gi protein-coupled receptors (SST1-5). Although its endocrine regulatory and anti-tumour effects have been thoroughly studied, little is known about its effect on the vascular system. The aim of the present study was to analyse the effects and potential mechanisms of somatostatin on endothelial barrier function. Cultured human umbilical vein endothelial cells (HUVECs) express mainly SST1 and SST5 receptors. Somatostatin did not affect the basal HUVEC permeability, but primed HUVEC monolayers for thrombin-induced hyperpermeability. Western blot data demonstrated that somatostatin activated the phosphoinositide 3-kinases (PI3K)/protein kinase B (Akt) and p42/44 mitogen-activated protein kinase (MAPK) pathways by phosphorylation. The HUVEC barrier destabilizing effects were abrogated by pre-treating HUVECs with mitogen-activated protein kinase kinase/extracellular signal regulated kinase (MEK/ERK), but not the Akt inhibitor. Moreover, somatostatin pre-treatment amplified vascular endothelial growth factor (VEGF)-induced angiogenesis (3D spheroid formation) in HUVECs. In conclusion, the data demonstrate that HUVECs under quiescence conditions express SST1 and SST5 receptors. Moreover, somatostatin primes HUVECs for thrombin-induced hyperpermeability mainly via the activation of MEK/ERK signalling and promotes HUVEC proliferation and angiogenesis in vitro.
Collapse
|
28
|
The Tyrosine Phosphatase hPTPRβ Controls the Early Signals and Dopaminergic Cells Viability via the P2X 7 Receptor. Int J Mol Sci 2021; 22:ijms222312936. [PMID: 34884741 PMCID: PMC8657974 DOI: 10.3390/ijms222312936] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 11/23/2021] [Accepted: 11/26/2021] [Indexed: 11/30/2022] Open
Abstract
ATP, one of the signaling molecules most commonly secreted in the nervous system and capable of stimulating multiple pathways, binds to the ionotropic purinergic receptors, in particular, the P2X7 receptor (P2X7R) and stimulates neuronal cell death. Given this effect of purinergic receptors on the viability of dopaminergic neurons model cells and that Ras GTPases control Erk1/2-regulated mitogen-activated cell proliferation and survival, we have investigated the role of the small GTPases of the Ras superfamily, together with their regulatory and effector molecules as the potential molecular intermediates in the P2X7R-regulated cell death of SN4741 dopaminergic neurons model cells. Here, we demonstrate that the neuronal response to purinergic stimulation involves the Calmodulin/RasGRF1 activation of the small GTPase Ras and Erk1/2. We also demonstrate that tyrosine phosphatase PTPRβ and other tyrosine phosphatases regulate the small GTPase activation pathway and neuronal viability. Our work expands the knowledge on the intracellular responses of dopaminergic cells by identifying new participating molecules and signaling pathways. In this sense, the study of the molecular circuitry of these neurons is key to understanding the functional effects of ATP, as well as considering the importance of these cells in Parkinson’s Disease.
Collapse
|
29
|
Amado-Azevedo J, van Stalborch AMD, Valent ET, Nawaz K, van Bezu J, Eringa EC, Hoevenaars FPM, De Cuyper IM, Hordijk PL, van Hinsbergh VWM, van Nieuw Amerongen GP, Aman J, Margadant C. Depletion of Arg/Abl2 improves endothelial cell adhesion and prevents vascular leak during inflammation. Angiogenesis 2021; 24:677-693. [PMID: 33770321 PMCID: PMC7996118 DOI: 10.1007/s10456-021-09781-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 03/06/2021] [Indexed: 02/06/2023]
Abstract
Endothelial barrier disruption and vascular leak importantly contribute to organ dysfunction and mortality during inflammatory conditions like sepsis and acute respiratory distress syndrome. We identified the kinase Arg/Abl2 as a mediator of endothelial barrier disruption, but the role of Arg in endothelial monolayer regulation and its relevance in vivo remain poorly understood. Here we show that depletion of Arg in endothelial cells results in the activation of both RhoA and Rac1, increased cell spreading and elongation, redistribution of integrin-dependent cell-matrix adhesions to the cell periphery, and improved adhesion to the extracellular matrix. We further show that Arg is activated in the endothelium during inflammation, both in murine lungs exposed to barrier-disruptive agents, and in pulmonary microvessels of septic patients. Importantly, Arg-depleted endothelial cells were less sensitive to barrier-disruptive agents. Despite the formation of F-actin stress fibers and myosin light chain phosphorylation, Arg depletion diminished adherens junction disruption and intercellular gap formation, by reducing the disassembly of cell-matrix adhesions and cell retraction. In vivo, genetic deletion of Arg diminished vascular leak in the skin and lungs, in the presence of a normal immune response. Together, our data indicate that Arg is a central and non-redundant regulator of endothelial barrier integrity, which contributes to cell retraction and gap formation by increasing the dynamics of adherens junctions and cell-matrix adhesions in a Rho GTPase-dependent fashion. Therapeutic inhibition of Arg may provide a suitable strategy for the treatment of a variety of clinical conditions characterized by vascular leak.
Collapse
Affiliation(s)
- Joana Amado-Azevedo
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | | | - Erik T Valent
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Kalim Nawaz
- Sanquin Research, Amsterdam, The Netherlands
| | - Jan van Bezu
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Etto C Eringa
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Femke P M Hoevenaars
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | | | - Peter L Hordijk
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Victor W M van Hinsbergh
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Geerten P van Nieuw Amerongen
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Jurjan Aman
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Amsterdam, The Netherlands. .,Department of Pulmonology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Amsterdam, The Netherlands.
| | - Coert Margadant
- Angiogenesis Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
30
|
Eskandarpour M, Nunn MA, Weston-Davies W, Calder VL. Immune-Mediated Retinal Vasculitis in Posterior Uveitis and Experimental Models: The Leukotriene (LT)B4-VEGF Axis. Cells 2021; 10:cells10020396. [PMID: 33671954 PMCID: PMC7919050 DOI: 10.3390/cells10020396] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/09/2021] [Accepted: 02/12/2021] [Indexed: 02/07/2023] Open
Abstract
Retinal vascular diseases have distinct, complex and multifactorial pathogeneses yet share several key pathophysiological aspects including inflammation, vascular permeability and neovascularisation. In non-infectious posterior uveitis (NIU), retinal vasculitis involves vessel leakage leading to retinal enlargement, exudation, and macular oedema. Neovascularisation is not a common feature in NIU, however, detection of the major angiogenic factor—vascular endothelial growth factor A (VEGF-A)—in intraocular fluids in animal models of uveitis may be an indication for a role for this cytokine in a highly inflammatory condition. Suppression of VEGF-A by directly targeting the leukotriene B4 (LTB4) receptor (BLT1) pathway indicates a connection between leukotrienes (LTs), which have prominent roles in initiating and propagating inflammatory responses, and VEGF-A in retinal inflammatory diseases. Further research is needed to understand how LTs interact with intraocular cytokines in retinal inflammatory diseases to guide the development of novel therapeutic approaches targeting both inflammatory mediator pathways.
Collapse
Affiliation(s)
- Malihe Eskandarpour
- UCL Institute of Ophthalmology, University College London, London EC1V 9EL, UK;
- Correspondence:
| | - Miles A. Nunn
- Akari Therapeutics Plc, London EC1V 9EL, UK; (M.A.N.); (W.W.-D.)
| | | | - Virginia L. Calder
- UCL Institute of Ophthalmology, University College London, London EC1V 9EL, UK;
| |
Collapse
|
31
|
Purinergic Regulation of Endothelial Barrier Function. Int J Mol Sci 2021; 22:ijms22031207. [PMID: 33530557 PMCID: PMC7865261 DOI: 10.3390/ijms22031207] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/10/2021] [Accepted: 01/22/2021] [Indexed: 12/12/2022] Open
Abstract
Increased vascular permeability is a hallmark of several cardiovascular anomalies, including ischaemia/reperfusion injury and inflammation. During both ischaemia/reperfusion and inflammation, massive amounts of various nucleotides, particularly adenosine 5'-triphosphate (ATP) and adenosine, are released that can induce a plethora of signalling pathways via activation of several purinergic receptors and may affect endothelial barrier properties. The nature of the effects on endothelial barrier function may depend on the prevalence and type of purinergic receptors activated in a particular tissue. In this review, we discuss the influence of the activation of various purinergic receptors and downstream signalling pathways on vascular permeability during pathological conditions.
Collapse
|
32
|
Chinigò G, Fiorio Pla A, Gkika D. TRP Channels and Small GTPases Interplay in the Main Hallmarks of Metastatic Cancer. Front Pharmacol 2020; 11:581455. [PMID: 33132914 PMCID: PMC7550629 DOI: 10.3389/fphar.2020.581455] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 09/11/2020] [Indexed: 12/13/2022] Open
Abstract
Transient Receptor Potential (TRP) cations channels, as key regulators of intracellular calcium homeostasis, play a central role in the essential hallmarks of cancer. Among the multiple pathways in which TRPs may be involved, here we focus our attention on the ones involving small guanosine triphosphatases (GTPases), summarizing the main processes associated with the metastatic cascade, such as migration, invasion and tumor vascularization. In the last decade, several studies have highlighted a bidirectional interplay between TRPs and small GTPases in cancer progression: TRP channels may affect small GTPases activity via both Ca2+-dependent or Ca2+-independent pathways, and, conversely, some small GTPases may affect TRP channels activity through the regulation of their intracellular trafficking to the plasma membrane or acting directly on channel gating. In particular, we will describe the interplay between TRPC1, TRPC5, TRPC6, TRPM4, TRPM7 or TRPV4, and Rho-like GTPases in regulating cell migration, the cooperation of TRPM2 and TRPV2 with Rho GTPases in increasing cell invasiveness and finally, the crosstalk between TRPC1, TRPC6, TRPM8, TRPV4 and both Rho- and Ras-like GTPases in inducing aberrant tumor vascularization.
Collapse
Affiliation(s)
- Giorgia Chinigò
- Laboratory of Cellular and Molecular Angiogenesis, Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy.,Laboratoire de Cell Physiology, Université de Lille, Department of Life Sciences, Univ. Lille, Inserm, U1003-PHYCEL, Lille, France
| | - Alessandra Fiorio Pla
- Laboratory of Cellular and Molecular Angiogenesis, Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy.,Laboratoire de Cell Physiology, Université de Lille, Department of Life Sciences, Univ. Lille, Inserm, U1003-PHYCEL, Lille, France
| | - Dimitra Gkika
- Laboratoire de Cell Physiology, Université de Lille, Department of Life Sciences, Univ. Lille, Inserm, U1003-PHYCEL, Lille, France.,Univ. Lille, CNRS, INSERM, CHU Lille, Centre Oscar Lambret, UMR 9020-UMR 1277-Canther-Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France.,Institut Universitaire de France (IUF), Paris, France
| |
Collapse
|
33
|
Yi L, Liang Y, Zhao Q, Wang H, Dong J. CX3CL1 Induces Vertebral Microvascular Barrier Dysfunction via the Src/P115-RhoGEF/ROCK Signaling Pathway. Front Cell Neurosci 2020; 14:96. [PMID: 32390803 PMCID: PMC7193116 DOI: 10.3389/fncel.2020.00096] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 03/31/2020] [Indexed: 12/12/2022] Open
Abstract
Trans-endothelial migration (TEM) of cancer cells is a critical step in metastasis. Micro-vascular barrier disruptions of distant organs play important roles in tumor cells TEM. The spine is a preferred site for multiple cancer cell metastases. Our previous study found that vertebral spongy bone was rich in CX3CL1 and that CX3CL1 can attract fractalkine receptor-expressing tumor cells to the spine. In the present study, we determined whether CX3CL1 was involved in vertebral micro-vascular barrier disruption and promoted tumor cell TEM after circulating tumor cells were arrested in the vertebral micro-vasculature. We examined the role of CX3CL1 in the barrier function of vertebral micro-vascular endothelial cells (VMECs) and explored the molecular mechanisms of CX3CL1-induced VMEC barrier disruption. Our results demonstrated that CX3CL1 led to F-actin formation and ZO-1 disruption in VMECs and induced the vertebral micro-vascular barrier disruption. Importantly, we found that the activation of the Src/P115-RhoGEF/ROCK signaling pathway plays an important role in CX3CL1-induced VMEC stress fiber formation, ZO-1 disruption and then vertebral micro-vascular barrier hyper-permeability. Inhibiting Src/P115-RhoGEF/ROCK signaling in VMECs effectively blocked CX3CL1-induced vertebral vascular endothelial dysfunction and subsequent tumor cell TEM. The results of this study and our previous study indicate that in addition to its chemotaxis, CX3CL1 plays a critical role in regulating vertebral micro-vascular barrier function and tumor cell TEM. CX3CL1 induced VMECs stress fiber formation, ZO-1 disruption and then vascular endothelial hyperpermeability via activation of the Src/P115-RhoGEF/ROCK signaling pathway. The inhibition of the Src/P115-RhoGEF/ROCK signaling pathway in VMECs effectively blocked tumor cells TEMs in vertebral spongy bone and maybe a potential therapeutic strategy for spine metastases in the future.
Collapse
Affiliation(s)
- Lei Yi
- Department of Burn and Plastic Surgery, School of Medicine, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, China.,Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yun Liang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Quanming Zhao
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Houlei Wang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jian Dong
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
34
|
Chaudhary PK, Han JS, Jee Y, Lee SH, Kim S. Pyk2 downstream of G 12/13 pathways regulates platelet shape change through RhoA/p160 ROCK. Biochem Biophys Res Commun 2020; 526:738-743. [PMID: 32265034 DOI: 10.1016/j.bbrc.2020.03.130] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/16/2020] [Accepted: 03/20/2020] [Indexed: 11/29/2022]
Abstract
Rho/Rho-kinase downstream of G12/13 plays an important role in the regulation of calcium-independent platelet shape change. We have previously shown that proline-rich tyrosine kinase 2 (Pyk2) is activated downstream of G12/13 pathways. In this study, we evaluated the role of Pyk2 in G12/13-induced platelet shape change. We used low concentrations of YFLLRNP, a heptapeptide binding to protease-activated receptor 1 (PAR1), or PAR4-activating peptide AYPGKF in the presence of Gαq inhibitor YM254890 to selectively stimulate G12/13 pathways. We found that G12/13-induced platelet shape change was completely inhibited in the presence of Pyk2 inhibitors AG17 and TAT-Pyk2-CT, suggesting an important role of Pyk2 in platelet shape change. In addition, AYPGKF-induced shape change in Gq -/- platelets was completely inhibited in the presence of AG17 or RhoA/p160ROCK inhibitor Y27632, confirming the role of Pyk2 in RhoA-dependent shape change. Furthermore, AYPGKF-induced platelet aggregation and dense granule secretion were inhibited by blocking Pyk2 or RhoA. Finally, G12/13-induced myosin phosphatase target subunit 1 (MYPT1) phosphorylation was inhibited by AG17, confirming that Pyk2 regulates RhoA/p160ROCK activation in platelets. These results demonstrate that Pyk2 downstream of G12/13 pathways regulates platelet shape change as well as platelet aggregation and dense granule secretion through the regulation of RhoA/p160ROCK.
Collapse
Affiliation(s)
| | - Jeung-Sul Han
- College of Agriculture & Life Sciences, Kyungpook National University, Daegu, 41566, South Korea
| | - Youngheun Jee
- College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju, 63243, South Korea
| | - Seung-Hun Lee
- College of Veterinary Medicine, Chungbuk National University, Cheongju, 28644, South Korea
| | - Soochong Kim
- College of Veterinary Medicine, Chungbuk National University, Cheongju, 28644, South Korea.
| |
Collapse
|
35
|
Tsuchiya T, Doi R, Obata T, Hatachi G, Nagayasu T. Lung Microvascular Niche, Repair, and Engineering. Front Bioeng Biotechnol 2020; 8:105. [PMID: 32154234 PMCID: PMC7047880 DOI: 10.3389/fbioe.2020.00105] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 02/03/2020] [Indexed: 12/28/2022] Open
Abstract
Biomaterials have been used for a long time in the field of medicine. Since the success of "tissue engineering" pioneered by Langer and Vacanti in 1993, tissue engineering studies have advanced from simple tissue generation to whole organ generation with three-dimensional reconstruction. Decellularized scaffolds have been widely used in the field of reconstructive surgery because the tissues used to generate decellularized scaffolds can be easily harvested from animals or humans. When a patient's own cells can be seeded onto decellularized biomaterials, theoretically this will create immunocompatible organs generated from allo- or xeno-organs. The most important aspect of lung tissue engineering is that the delicate three-dimensional structure of the organ is maintained during the tissue engineering process. Therefore, organ decellularization has special advantages for lung tissue engineering where it is essential to maintain the extremely thin basement membrane in the alveoli. Since 2010, there have been many methodological developments in the decellularization and recellularization of lung scaffolds, which includes improvements in the decellularization protocols and the selection and preparation of seeding cells. However, early transplanted engineered lungs terminated in organ failure in a short period. Immature vasculature reconstruction is considered to be the main cause of engineered organ failure. Immature vasculature causes thrombus formation in the engineered lung. Successful reconstruction of a mature vasculature network would be a major breakthrough in achieving success in lung engineering. In order to regenerate the mature vasculature network, we need to remodel the vascular niche, especially the microvasculature, in the organ scaffold. This review highlights the reconstruction of the vascular niche in a decellularized lung scaffold. Because the vascular niche consists of endothelial cells (ECs), pericytes, extracellular matrix (ECM), and the epithelial-endothelial interface, all of which might affect the vascular tight junction (TJ), we discuss ECM composition and reconstruction, the contribution of ECs and perivascular cells, the air-blood barrier (ABB) function, and the effects of physiological factors during the lung microvasculature repair and engineering process. The goal of the present review is to confirm the possibility of success in lung microvascular engineering in whole organ engineering and explore the future direction of the current methodology.
Collapse
Affiliation(s)
- Tomoshi Tsuchiya
- Department of Surgical Oncology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan.,Division of Nucleic Acid Drug Development, Research Institute for Science and Technology, Tokyo University of Science, Chiba, Japan
| | - Ryoichiro Doi
- Department of Surgical Oncology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Tomohiro Obata
- Department of Surgical Oncology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Go Hatachi
- Department of Surgical Oncology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Takeshi Nagayasu
- Department of Surgical Oncology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
36
|
MLCK and ROCK mutualism in endothelial barrier dysfunction. Biochimie 2020; 168:83-91. [DOI: 10.1016/j.biochi.2019.10.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 10/22/2019] [Indexed: 01/15/2023]
|
37
|
Gündüz D, Troidl C, Tanislav C, Rohrbach S, Hamm C, Aslam M. Role of PI3K/Akt and MEK/ERK Signalling in cAMP/Epac-Mediated Endothelial Barrier Stabilisation. Front Physiol 2019; 10:1387. [PMID: 31787905 PMCID: PMC6855264 DOI: 10.3389/fphys.2019.01387] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 10/22/2019] [Indexed: 12/16/2022] Open
Abstract
Background and Aims Activation of the cAMP/Epac signalling stabilises endothelial barrier function. Moreover, its activation is accompanied by an activation of PI3K/Akt and MEK/ERK signalling in diverse cell types but their impact on endothelial barrier function is largely unknown. Here the role of PI3K/Akt and MEK/ERK signalling in cAMP/Epac-mediated endothelial barrier stabilisation was analysed. Methods Endothelial barrier function was analysed in cultured human umbilical vein endothelial cells (HUVECs) by measuring flux of albumin. A modified cAMP analogue 8-pCPT-2′-O-Me-cAMP (Epac agonist) was used to specifically activate cAMP/Epac signalling. Results Epac agonist reduces the basal and attenuates thrombin-induced endothelial hyperpermeability accompanied by an activation of PI3K/Akt and MEK/ERK signalling. The qPCR data demonstrate HUVECs express PI3Kα, PI3Kβ, and PI3Kγ but not PI3Kδ isoforms. The western blot data demonstrate Epac agonist activates PI3Kα and PI3Kβ isoforms. Inhibition of MEK/ERK but not PI3K/Akt pathway potentiates the endothelial barrier protective effects of cAMP/Epac signalling. Inhibition of MEK/ERK signalling in the presence of Epac agonist induces a reorganisation of actin cytoskeleton to the cell periphery, enhanced VE-cadherin localisation at cell-cell junctions, and dephosphorylation of myosin light chains (MLC) but not inhibition of RhoA/Rock signalling. Moreover, Epac agonist promotes endothelial cell (EC) survival via reduction in activities of pro-apoptotic caspases in a PI3K/Akt and MEK/ERK signalling-dependent manner. Conclusion Our data demonstrate that the Epac agonist simultaneously activates diverse signalling pathways in ECs, which may have differential effects on endothelial barrier function. It activates PI3K/Akt and MEK/ERK signalling which mainly govern its pro-survival effects on ECs. Inhibition of MEK/ERK but not PI3K/Akt signalling enhances barrier stabilising and barrier protective effects of cAMP/Epac activation. Chemical Compounds Used In This Study 8-pCPT-2′-O-Me-cAMP (PubChem CID: 9913268); Akt inhibitor VIII (PubChem CID: 10196499); AS-252424 (PubChem CID: 11630874); IC-87114 (PubChem CID: 9908783); PD 98059 (PubChem CID: 4713); PIK-75 (PubChem CID: 10275789); TGX-221 (PubChem CID: 9907093); Thrombin (PubChem CID: 90470996); U0126 (PubChem CID: 3006531); Wortmannin (PubChem CID: 312145).
Collapse
Affiliation(s)
- Dursun Gündüz
- Department of Cardiology and Angiology, University Hospital of Giessen and Marburg, Giessen, Germany.,Department of Cardiology and Angiology, Evangelisches Jung Stilling Krankenhaus GmbH, Siegen, Germany
| | - Christian Troidl
- Department of Cardiology and Angiology, University Hospital of Giessen and Marburg, Giessen, Germany.,Experimental Cardiology, Justus Liebig University Giessen, Giessen, Germany
| | - Christian Tanislav
- Department of Neurology, Evangelisches Jung Stilling Krankenhaus GmbH, Siegen, Germany.,Department of Neurology, University Hospital of Giessen and Marburg, Giessen, Germany
| | - Susanne Rohrbach
- Institute of Physiology, Justus Liebig University Giessen, Giessen, Germany
| | - Christian Hamm
- Department of Cardiology and Angiology, University Hospital of Giessen and Marburg, Giessen, Germany
| | - Muhammad Aslam
- Department of Cardiology and Angiology, University Hospital of Giessen and Marburg, Giessen, Germany.,Experimental Cardiology, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
38
|
Karki P, Birukov KG. Rho and Reactive Oxygen Species at Crossroads of Endothelial Permeability and Inflammation. Antioxid Redox Signal 2019; 31:1009-1022. [PMID: 31126187 PMCID: PMC6765062 DOI: 10.1089/ars.2019.7798] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significance: Increased endothelial permeability and inflammation are two major hallmarks of the life-threatening conditions such as acute respiratory distress syndrome and sepsis. There is a growing consensus in the field that the Rho family of small guanosine triphosphates are critical regulators of endothelial function at both physiological and pathological states. A basal level of reactive oxygen species (ROS) is essential for maintaining metabolic homeostasis, vascular tone, and angiogenesis; however, excessive ROS generation impairs endothelial function and promotes lung inflammation. In this review, we will focus on the role of Rho in control of endothelial function and also briefly discuss a nexus between ROS generation and Rho activation during endothelial dysfunction. Recent Advances: Extensive studies in the past decades have established that a wide range of barrier-disruptive and proinflammatory agonists activate the Rho pathway that, ultimately, leads to endothelial dysfunction via disruption of endothelial barrier and further escalation of inflammation. An increasing body of evidence suggests that a bidirectional interplay exists between the Rho pathway and ROS generation during endothelial dysfunction. Rac, a member of the Rho family, is directly involved in ROS production and ROS, in turn, activate RhoA, Rac, and Cdc42. Critical Issues: A precise mechanism of interaction between ROS generation and Rho activation and its impact on endothelial function needs to be elucidated. Future Directions: By employing advanced molecular techniques, the sequential cascades in the Rho-ROS crosstalk signaling axis need to be explored. The therapeutic potential of the Rho pathway inhibitors in endothelial-dysfunction associated cardiopulmonary disorders needs to be evaluated.
Collapse
Affiliation(s)
- Pratap Karki
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland, Baltimore, Maryland
| | - Konstantin G Birukov
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
39
|
Wettschureck N, Strilic B, Offermanns S. Passing the Vascular Barrier: Endothelial Signaling Processes Controlling Extravasation. Physiol Rev 2019; 99:1467-1525. [PMID: 31140373 DOI: 10.1152/physrev.00037.2018] [Citation(s) in RCA: 188] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
A central function of the vascular endothelium is to serve as a barrier between the blood and the surrounding tissue of the body. At the same time, solutes and cells have to pass the endothelium to leave or to enter the bloodstream to maintain homeostasis. Under pathological conditions, for example, inflammation, permeability for fluid and cells is largely increased in the affected area, thereby facilitating host defense. To appropriately function as a regulated permeability filter, the endothelium uses various mechanisms to allow solutes and cells to pass the endothelial layer. These include transcellular and paracellular pathways of which the latter requires remodeling of intercellular junctions for its regulation. This review provides an overview on endothelial barrier regulation and focuses on the endothelial signaling mechanisms controlling the opening and closing of paracellular pathways for solutes and cells such as leukocytes and metastasizing tumor cells.
Collapse
Affiliation(s)
- Nina Wettschureck
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| | - Boris Strilic
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| |
Collapse
|
40
|
Yoon C, Choi C, Stapleton S, Mirabella T, Howes C, Dong L, King J, Yang J, Oberai A, Eyckmans J, Chen CS. Myosin IIA-mediated forces regulate multicellular integrity during vascular sprouting. Mol Biol Cell 2019; 30:1974-1984. [PMID: 31318321 PMCID: PMC6727772 DOI: 10.1091/mbc.e19-02-0076] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Angiogenic sprouting is a critical process involved in vascular network formation within tissues. During sprouting, tip cells and ensuing stalk cells migrate collectively into the extracellular matrix while preserving cell–cell junctions, forming patent structures that support blood flow. Although several signaling pathways have been identified as controlling sprouting, it remains unclear to what extent this process is mechanoregulated. To address this question, we investigated the role of cellular contractility in sprout morphogenesis, using a biomimetic model of angiogenesis. Three-dimensional maps of mechanical deformations generated by sprouts revealed that mainly leader cells, not stalk cells, exert contractile forces on the surrounding matrix. Surprisingly, inhibiting cellular contractility with blebbistatin did not affect the extent of cellular invasion but resulted in cell–cell dissociation primarily between tip and stalk cells. Closer examination of cell–cell junctions revealed that blebbistatin impaired adherens-junction organization, particularly between tip and stalk cells. Using CRISPR/Cas9-mediated gene editing, we further identified NMIIA as the major isoform responsible for regulating multicellularity and cell contractility during sprouting. Together, these studies reveal a critical role for NMIIA-mediated contractile forces in maintaining multicellularity during sprouting and highlight the central role of forces in regulating cell–cell adhesions during collective motility.
Collapse
Affiliation(s)
- Christine Yoon
- Biological Design Center, Department of Biomedical Engineering, Boston University, Boston, MA 02215
| | - Colin Choi
- Biological Design Center, Department of Biomedical Engineering, Boston University, Boston, MA 02215
| | - Sarah Stapleton
- Biological Design Center, Department of Biomedical Engineering, Boston University, Boston, MA 02215
| | - Teodelinda Mirabella
- Biological Design Center, Department of Biomedical Engineering, Boston University, Boston, MA 02215
| | - Caroline Howes
- Department of Mechanical, Aerospace and Nuclear Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180
| | - Li Dong
- Department of Mechanical, Aerospace and Nuclear Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180.,The Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, TX 78712
| | - Jessica King
- Biological Design Center, Department of Biomedical Engineering, Boston University, Boston, MA 02215
| | - Jinling Yang
- Biological Design Center, Department of Biomedical Engineering, Boston University, Boston, MA 02215
| | - Assad Oberai
- Department of Mechanical, Aerospace and Nuclear Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180.,Department of Aerospace and Mechanical Engineering, University of Southern California, Los Angeles, CA 90007
| | - Jeroen Eyckmans
- Biological Design Center, Department of Biomedical Engineering, Boston University, Boston, MA 02215.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115
| | - Christopher S Chen
- Biological Design Center, Department of Biomedical Engineering, Boston University, Boston, MA 02215.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115
| |
Collapse
|
41
|
Shakhov AS, Dugina VB, Alieva IB. Structural Features of Actin Cytoskeleton Required for Endotheliocyte Barrier Function. BIOCHEMISTRY (MOSCOW) 2019; 84:358-369. [PMID: 31228927 DOI: 10.1134/s0006297919040035] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Cytoplasmic actin structures are essential components of the eukaryotic cytoskeleton. According to the classic concepts, actin structures perform contractile and motor functions, ensuring the possibility of cell shape changes during cell spreading, polarization, and movement both in vitro and in vivo, from the early embryogenesis stages and throughout the life of a multicellular organism. Intracellular organization of actin structures, their biochemical composition, and dynamic properties play a key role in the realization of specific cellular and tissue functions and vary in different cell types. This paper is a review of recent studies on the organization and properties of actin structures in endotheliocytes, interaction of these structures with other cytoskeletal components and elements involved in cell adhesion, as well as their role in the functional activity of endothelial cells.
Collapse
Affiliation(s)
- A S Shakhov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119992, Russia
| | - V B Dugina
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119992, Russia
| | - I B Alieva
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119992, Russia.
| |
Collapse
|
42
|
Stiff Substrates Enhance Endothelial Oxidative Stress in Response to Protein Kinase C Activation. Appl Bionics Biomech 2019; 2019:6578492. [PMID: 31110559 PMCID: PMC6487160 DOI: 10.1155/2019/6578492] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 01/28/2019] [Accepted: 02/19/2019] [Indexed: 02/07/2023] Open
Abstract
Arterial stiffness, which increases with aging and hypertension, is an independent cardiovascular risk factor. While stiffer substrates are known to affect single endothelial cell morphology and migration, the effect of substrate stiffness on endothelial monolayer function is less understood. The objective of this study was to determine if substrate stiffness increased endothelial monolayer reactive oxygen species (ROS) in response to protein kinase C (PKC) activation and if this oxidative stress then impacted adherens junction integrity. Porcine aortic endothelial cells were cultured on varied stiffness polyacrylamide gels and treated with phorbol 12-myristate 13-acetate (PMA), which stimulates PKC and ROS without increasing actinomyosin contractility. PMA-treated endothelial cells on stiffer substrates increased ROS and adherens junction loss without increased contractility. ROS scavengers abrogated PMA effects on cell-cell junctions, with a more profound effect in cells on stiffer substrates. Finally, endothelial cells in aortae from elastin haploinsufficient mice (Eln+/-), which were stiffer than aortae from wild-type mice, showed decreased VE-cadherin colocalization with peripheral actin following PMA treatment. These data suggest that oxidative stress may be enhanced in endothelial cells in stiffer vessels, which could contribute to the association between arterial stiffness and cardiovascular disease.
Collapse
|
43
|
Ke Y, Karki P, Zhang C, Li Y, Nguyen T, Birukov KG, Birukova AA. Mechanosensitive Rap1 activation promotes barrier function of lung vascular endothelium under cyclic stretch. Mol Biol Cell 2019; 30:959-974. [PMID: 30759056 PMCID: PMC6589902 DOI: 10.1091/mbc.e18-07-0422] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Mechanical ventilation remains an imperative treatment for the patients with acute respiratory distress syndrome, but can also exacerbate lung injury. We have previously described a key role of RhoA GTPase in high cyclic stretch (CS)-induced endothelial cell (EC) barrier dysfunction. However, cellular mechanotransduction complexes remain to be characterized. This study tested a hypothesis that recovery of a vascular EC barrier after pathologic mechanical stress may be accelerated by cell exposure to physiologic CS levels and involves Rap1-dependent rearrangement of endothelial cell junctions. Using biochemical, molecular, and imaging approaches we found that EC pre- or postconditioning at physiologically relevant low-magnitude CS promotes resealing of cell junctions disrupted by pathologic, high-magnitude CS. Cytoskeletal remodeling induced by low CS was dependent on small GTPase Rap1. Protective effects of EC preconditioning at low CS were abolished by pharmacological or molecular inhibition of Rap1 activity. In vivo, using mice exposed to mechanical ventilation, we found that the protective effect of low tidal volume ventilation against lung injury caused by lipopolysaccharides and ventilation at high tidal volume was suppressed in Rap1 knockout mice. Taken together, our results demonstrate a prominent role of Rap1-mediated signaling mechanisms activated by low CS in acceleration of lung vascular EC barrier restoration.
Collapse
Affiliation(s)
- Yunbo Ke
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Pratap Karki
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Chenou Zhang
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Yue Li
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Trang Nguyen
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Konstantin G. Birukov
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Anna A. Birukova
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201,*Address correspondence to: Anna A. Birukova ()
| |
Collapse
|
44
|
Fang Y, Wu D, Birukov KG. Mechanosensing and Mechanoregulation of Endothelial Cell Functions. Compr Physiol 2019; 9:873-904. [PMID: 30873580 PMCID: PMC6697421 DOI: 10.1002/cphy.c180020] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Vascular endothelial cells (ECs) form a semiselective barrier for macromolecules and cell elements regulated by dynamic interactions between cytoskeletal elements and cell adhesion complexes. ECs also participate in many other vital processes including innate immune reactions, vascular repair, secretion, and metabolism of bioactive molecules. Moreover, vascular ECs represent a unique cell type exposed to continuous, time-dependent mechanical forces: different patterns of shear stress imposed by blood flow in macrovasculature and by rolling blood cells in the microvasculature; circumferential cyclic stretch experienced by the arterial vascular bed caused by heart propulsions; mechanical stretch of lung microvascular endothelium at different magnitudes due to spontaneous respiration or mechanical ventilation in critically ill patients. Accumulating evidence suggests that vascular ECs contain mechanosensory complexes, which rapidly react to changes in mechanical loading, process the signal, and develop context-specific adaptive responses to rebalance the cell homeostatic state. The significance of the interactions between specific mechanical forces in the EC microenvironment together with circulating bioactive molecules in the progression and resolution of vascular pathologies including vascular injury, atherosclerosis, pulmonary edema, and acute respiratory distress syndrome has been only recently recognized. This review will summarize the current understanding of EC mechanosensory mechanisms, modulation of EC responses to humoral factors by surrounding mechanical forces (particularly the cyclic stretch), and discuss recent findings of magnitude-specific regulation of EC functions by transcriptional, posttranscriptional and epigenetic mechanisms using -omics approaches. We also discuss ongoing challenges and future opportunities in developing new therapies targeting dysregulated mechanosensing mechanisms to treat vascular diseases. © 2019 American Physiological Society. Compr Physiol 9:873-904, 2019.
Collapse
Affiliation(s)
- Yun Fang
- Department of Medicine, University of Chicago, Chicago, Illinois, USA,Correspondence to
| | - David Wu
- Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Konstantin G. Birukov
- Department of Anesthesiology, University of Maryland Baltimore School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
45
|
Flentje A, Kalsi R, Monahan TS. Small GTPases and Their Role in Vascular Disease. Int J Mol Sci 2019; 20:ijms20040917. [PMID: 30791562 PMCID: PMC6413073 DOI: 10.3390/ijms20040917] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 01/31/2019] [Accepted: 02/07/2019] [Indexed: 12/18/2022] Open
Abstract
Over eighty million people in the United States have cardiovascular disease that can affect the heart causing myocardial infarction; the carotid arteries causing stroke; and the lower extremities leading to amputation. The treatment for end-stage cardiovascular disease is surgical—either endovascular therapy with balloons and stents—or open reconstruction to reestablish blood flow. All interventions damage or destroy the protective inner lining of the blood vessel—the endothelium. An intact endothelium is essential to provide a protective; antithrombotic lining of a blood vessel. Currently; there are no agents used in the clinical setting that promote reendothelialization. This process requires migration of endothelial cells to the denuded vessel; proliferation of endothelial cells on the denuded vessel surface; and the reconstitution of the tight adherence junctions responsible for the formation of an impermeable surface. These processes are all regulated in part and are dependent on small GTPases. As important as the small GTPases are for reendothelialization, dysregulation of these molecules can result in various vascular pathologies including aneurysm formation, atherosclerosis, diabetes, angiogenesis, and hypertension. A better understanding of the role of small GTPases in endothelial cell migration is essential to the development for novel agents to treat vascular disease.
Collapse
Affiliation(s)
- Alison Flentje
- Division of Vascular Surgery, Department of Surgery, University of Maryland School of Medicine, 22 South Greene Street, Suite S10B00, Baltimore, MD 21201, USA.
| | - Richa Kalsi
- Division of Vascular Surgery, Department of Surgery, University of Maryland School of Medicine, 22 South Greene Street, Suite S10B00, Baltimore, MD 21201, USA.
| | - Thomas S Monahan
- Division of Vascular Surgery, Department of Surgery, University of Maryland School of Medicine, 22 South Greene Street, Suite S10B00, Baltimore, MD 21201, USA.
| |
Collapse
|
46
|
Karki P, Ke Y, Tian Y, Ohmura T, Sitikov A, Sarich N, Montgomery CP, Birukova AA. Staphylococcus aureus-induced endothelial permeability and inflammation are mediated by microtubule destabilization. J Biol Chem 2019; 294:3369-3384. [PMID: 30622143 DOI: 10.1074/jbc.ra118.004030] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 12/13/2018] [Indexed: 12/18/2022] Open
Abstract
Staphylococcus aureus is a major etiological agent of sepsis and induces endothelial cell (EC) barrier dysfunction and inflammation, two major hallmarks of acute lung injury. However, the molecular mechanisms of bacterial pathogen-induced EC barrier disruption are incompletely understood. Here, we investigated the role of microtubules (MT) in the mechanisms of EC barrier compromise caused by heat-killed S. aureus (HKSA). Using a customized monolayer permeability assay in human pulmonary EC and MT fractionation, we observed that HKSA-induced barrier disruption is accompanied by MT destabilization and increased histone deacetylase-6 (HDAC6) activity resulting from elevated reactive oxygen species (ROS) production. Molecular or pharmacological HDAC6 inhibition rescued barrier function in HKSA-challenged vascular endothelium. The HKSA-induced EC permeability was associated with impaired MT-mediated delivery of cytoplasmic linker-associated protein 2 (CLASP2) to the cell periphery, limiting its interaction with adherens junction proteins. HKSA-induced EC barrier dysfunction was also associated with increased Rho GTPase activity via activation of MT-bound Rho-specific guanine nucleotide exchange factor-H1 (GEF-H1) and was abolished by HDAC6 down-regulation. HKSA activated the NF-κB proinflammatory pathway and increased the expression of intercellular and vascular cell adhesion molecules in EC, an effect that was also HDAC6-dependent and mediated, at least in part, by a GEF-H1/Rho-dependent mechanism. Of note, HDAC6 knockout mice or HDAC6 inhibitor-treated WT mice were partially protected from vascular leakage and inflammation caused by both HKSA or methicillin-resistant S. aureus (MRSA). Our results indicate that S. aureus-induced, ROS-dependent up-regulation of HDAC6 activity destabilizes MT and thereby activates the GEF-H1/Rho pathway, increasing both EC permeability and inflammation.
Collapse
Affiliation(s)
- Pratap Karki
- From the Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Yunbo Ke
- the Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Yufeng Tian
- the Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, Illinois 60637, and
| | - Tomomi Ohmura
- the Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, Illinois 60637, and
| | - Albert Sitikov
- the Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, Illinois 60637, and
| | - Nicolene Sarich
- the Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, Illinois 60637, and
| | - Christopher P Montgomery
- the Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, Illinois 60637, and.,the Department of Critical Care Medicine, Nationwide Children's Hospital, Columbus, Ohio 43205
| | - Anna A Birukova
- From the Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201,
| |
Collapse
|
47
|
Yin J, Lv L, Zhai P, Long T, Zhou Q, Pan H, Botwe G, Wang L, Wang Q, Tan L, Kuebler WM. Connexin 40 regulates lung endothelial permeability in acute lung injury via the ROCK1-MYPT1- MLC20 pathway. Am J Physiol Lung Cell Mol Physiol 2019; 316:L35-L44. [PMID: 30234377 DOI: 10.1152/ajplung.00012.2018] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Increased pulmonary vascular permeability is a hallmark of acute lung injury (ALI). Connexin 40 (Cx40) is a gap junctional protein abundantly present in the lung microvascular endothelium. Yet, the role of Cx40 in the regulation of lung vascular permeability and its underlying mechanisms are unclear. Here, we tested the hypothesis that Cx40 participates in regulation of lung endothelial permeability via a mechanism involving a Rho-associated protein kinase (ROCK) dependent regulation of myosin light chain (MLC). In murine models of intratracheal acid- or LPS-induced lung injury, genetic deficiency of Cx40 attenuated key features of ALI including vascular barrier failure. In human pulmonary microvascular endothelial cells (PMVECs), thrombin-induced loss of transendothelial electrical resistance was attenuated by a Cx40-inhibiting mimetic peptide (40GAP27), Cx40-specific shRNA, or ROCK inhibitor Y27632. In isolated perfused mouse lungs, platelet-activating factor-induced lung weight gain was abrogated by gap junction blocker carbenoxolone, 40GAP27, Y27632, or genetic deficiency of Cx40. Phosphorylation of MLC20 increased drastically in both LPS-treated PMVECs and HCl-treated mouse lungs. Expression of ROCK1 was increased in both LPS-treated PMVECs and HCl-treated mouse lungs, and paralleled by phosphorylation of MLC20. Coimmunoprecipitation experiments revealed protein-protein interaction between ROCK1 and Cx40. LPS-induced upregulation of ROCK1 and phosphorylation of MLC20 were blocked by knockdown of Cx40. LPS caused phosphorylation of myosin phosphatase targeting subunit 1, which could be abrogated by Y27632 or Cx40-shRNA. Our findings reveal a role of Cx40 in regulation of ROCK1 and MLC20 that contributes critically to lung vascular barrier failure in ALI.
Collapse
Affiliation(s)
- Jun Yin
- Department of Thoracic Surgery, Zhongshan Hospital of Fudan University , Shanghai , China
- Department of Cardiothoracic Surgery, Affiliated People's Hospital of Jiangsu University , Zhenjiang, Jiangsu , China
| | - Lu Lv
- Department of Cardiothoracic Surgery, Affiliated People's Hospital of Jiangsu University , Zhenjiang, Jiangsu , China
| | - Peng Zhai
- Department of Cardiothoracic Surgery, Affiliated People's Hospital of Jiangsu University , Zhenjiang, Jiangsu , China
| | - Tao Long
- Department of Cardiothoracic Surgery, Affiliated People's Hospital of Jiangsu University , Zhenjiang, Jiangsu , China
| | - Qiang Zhou
- Department of Cardiothoracic Surgery, Affiliated People's Hospital of Jiangsu University , Zhenjiang, Jiangsu , China
| | - Huiwen Pan
- Department of Cardiothoracic Surgery, Affiliated People's Hospital of Jiangsu University , Zhenjiang, Jiangsu , China
| | - Godwin Botwe
- Department of Cardiothoracic Surgery, Affiliated People's Hospital of Jiangsu University , Zhenjiang, Jiangsu , China
| | - Liming Wang
- Department of Chemotherapy, Cancer Institute, Affiliated People's Hospital of Jiangsu University , Zhenjiang, Jiangsu , China
| | - Qun Wang
- Department of Thoracic Surgery, Zhongshan Hospital of Fudan University , Shanghai , China
| | - Lijie Tan
- Department of Thoracic Surgery, Zhongshan Hospital of Fudan University , Shanghai , China
| | - Wolfgang M Kuebler
- Department of Physiology and Center for Cardiovascular Research, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health , Berlin , Germany
| |
Collapse
|
48
|
Endothelial Protrusions in Junctional Integrity and Barrier Function. CURRENT TOPICS IN MEMBRANES 2018; 82:93-140. [PMID: 30360784 DOI: 10.1016/bs.ctm.2018.08.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Endothelial cells of the microcirculation form a semi-permeable diffusion barrier between the blood and tissues. This permeability of the endothelium, particularly in the capillaries and postcapillary venules, is a normal physiological function needed for blood-tissue exchange in the microcirculation. During inflammation, microvascular permeability increases dramatically and can lead to tissue edema, which in turn can lead to dysfunction of tissues and organs. The molecular mechanisms that control the barrier function of endothelial cells have been under investigation for several decades and remain an important topic due to the potential for discovery of novel therapeutic strategies to reduce edema. This review highlights current knowledge of the cellular and molecular mechanisms that lead to endothelial hyperpermeability during inflammatory conditions associated with injury and disease. This includes a discussion of recent findings demonstrating temporal protrusions by endothelial cells that may contribute to intercellular junction integrity between endothelial cells and affect the diffusion distance for solutes via the paracellular pathway.
Collapse
|
49
|
Kiss A, Erdődi F, Lontay B. Myosin phosphatase: Unexpected functions of a long-known enzyme. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1866:2-15. [PMID: 30076859 DOI: 10.1016/j.bbamcr.2018.07.023] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 07/09/2018] [Accepted: 07/26/2018] [Indexed: 01/08/2023]
Abstract
Myosin phosphatase (MP) holoenzyme is a Ser/Thr specific enzyme, which is the member of protein phosphatase type 1 (PP1) family and composed of a PP1 catalytic subunit (PP1c/PPP1CB) and a myosin phosphatase targeting subunit (MYPT1/PPP1R12A). PP1c is required for the catalytic activity of the holoenzyme, while MYPT1 regulates MP through targeting the holoenzyme to its substrates. Above the well-characterized function of MP, as the major regulator of smooth muscle contractility mediating the dephosphorylation of 20 kDa myosin light chain, accumulating data support its role in other, non-contractile functions. In this review, we summarize the scaffold function of MP holoenzyme and its roles in processes such as cell cycle, development, gene expression regulation and neurotransmitter release. In particular, we highlight novel interacting proteins of MYPT1 and pathophysiological functions of MP relevant to tumorigenesis, insulin resistance and neurodegenerative disorders. This article is part of a Special Issue entitled: Protein Phosphatases as Critical Regulators for Cellular Homeostasis edited by Prof. Peter Ruvolo and Dr. Veerle Janssens.
Collapse
Affiliation(s)
- Andrea Kiss
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ferenc Erdődi
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; MTA-DE Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Beáta Lontay
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
50
|
Chen B, Yang Z, Yang C, Qin W, Gu J, Hu C, Chen A, Ning J, Yi B, Lu K. A self-organized actomyosin drives multiple intercellular junction disruption and directly promotes neutrophil recruitment in lipopolysaccharide-induced acute lung injury. FASEB J 2018; 32:fj201701506RR. [PMID: 29879372 DOI: 10.1096/fj.201701506rr] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Acute lung injury (ALI), with the hallmarks of vascular integrity disruption and neutrophil recruitment, is associated with high morbidity and mortality. Enhanced actomyosin assembly contributes to endothelial cell contact dysfunction. However, the roles and mechanisms of actomyosin assembly in ALI are not totally clear. We investigated the dynamic alterations and roles of actomyosin in ALI in vivo and in vitro models induced by LPS. Pulmonary levels of E-cadherin, vascular endothelial-cadherin, occludin, myosin phosphatase target subunit 1, and thymosin β4 were decreased, and the number and activity of neutrophils and the levels of actomyosin, p-ρ-associated protein kinase, p-myosin light-chain kinase, and profilin1 were increased within 3 d after LPS administration, and then, those alterations were recovered within the next 4 d, which was consistent with the alterations of lung histology, vascular permeability, edema, and serum levels of IL-6 and TNF-α. Direct or indirect inhibition of increased F-actin or myosin assembly ameliorated the reduction of intercellular junction molecules, the activation and migration of neutrophils, and the degree of lung injury. Moreover, neutrophil activation further promoted actomyosin assembly and aggravated lung injury. Conclusively, the enhancement of self-organized actomyosin contributes to alveolar-capillary barrier disruption and neutrophil recruitment in inflammatory response, which is a potential therapeutic target for ALI.-Chen, B., Yang, Z., Yang, C., Qin, W., Gu, J., Hu, C., Chen, A., Ning, J., Yi, B., Lu, K. A self-organized actomyosin drives multiple intercellular junction disruption and directly promotes neutrophil recruitment in lipopolysaccharide-induced acute lung injury.
Collapse
Affiliation(s)
- Bing Chen
- Department of Anesthesia, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Zhen Yang
- Department of Anesthesia, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Congwen Yang
- Department of Anesthesia, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Wenhan Qin
- Department of Anesthesia, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jianteng Gu
- Department of Anesthesia, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Chuanmin Hu
- Department of Clinical Biochemistry, College of Medical Laboratory, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - An Chen
- Department of Clinical Biochemistry, College of Medical Laboratory, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jiaolin Ning
- Department of Anesthesia, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Bin Yi
- Department of Anesthesia, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Kaizhi Lu
- Department of Anesthesia, Southwest Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|