1
|
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
|
2
|
Noone D, Preston RJS, Rehill AM. The Role of Myeloid Cells in Thromboinflammatory Disease. Semin Thromb Hemost 2024; 50:998-1011. [PMID: 38547918 DOI: 10.1055/s-0044-1782660] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Inflammation contributes to the development of thrombosis, but the mechanistic basis for this association remains poorly understood. Innate immune responses and coagulation pathways are activated in parallel following infection or injury, and represent an important host defense mechanism to limit pathogen spread in the bloodstream. However, dysregulated proinflammatory activity is implicated in the progression of venous thromboembolism and arterial thrombosis. In this review, we focus on the role of myeloid cells in propagating thromboinflammation in acute inflammatory conditions, such as sepsis and coronavirus disease 2019 (COVID-19), and chronic inflammatory conditions, such as obesity, atherosclerosis, and inflammatory bowel disease. Myeloid cells are considered key drivers of thromboinflammation via upregulated tissue factor activity, formation of neutrophil extracellular traps (NETs), contact pathway activation, and aberrant coagulation factor-mediated protease-activated receptor (PAR) signaling. We discuss how strategies to target the intersection between myeloid cell-mediated inflammation and activation of blood coagulation represent an exciting new approach to combat immunothrombosis. Specifically, repurposed anti-inflammatory drugs, immunometabolic regulators, and NETosis inhibitors present opportunities that have the potential to dampen immunothrombotic activity without interfering with hemostasis. Such therapies could have far-reaching benefits for patient care across many thromboinflammatory conditions.
Collapse
Affiliation(s)
- David Noone
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- National Children's Research Centre, Our Lady's Children's Hospital Crumlin, Dublin, Ireland
| | - Roger J S Preston
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- National Children's Research Centre, Our Lady's Children's Hospital Crumlin, Dublin, Ireland
| | - Aisling M Rehill
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- National Children's Research Centre, Our Lady's Children's Hospital Crumlin, Dublin, Ireland
| |
Collapse
|
3
|
Tao W, Min S, Chen G, He X, Meng Y, Li L, Chen J, Li Y. Tetramethylpyrazine ameliorates LPS-induced acute lung injury via the miR-369-3p/DSTN axis. Sci Rep 2024; 14:20006. [PMID: 39198493 PMCID: PMC11358269 DOI: 10.1038/s41598-024-70131-0] [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: 05/14/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024] Open
Abstract
Acute lung injury (ALI) is a severe clinical respiratory condition characterized by high rates of mortality and morbidity, for which effective treatments are currently lacking. In this study, lipopolysaccharide (LPS) was used to induce ALI mice, demonstrating the efficacy of tetramethylpyrazine (TMP) in ameliorating ALI. Subsequent we perfored high-throughput sequencing analysis and used Targetscan 8.0 and miRWalk 3.0 databases to predict the interaction between microRNAs and destrin (DSTN), ultimately identifying miR-369-3p as the focus of the investigation. The adenovirus carrying miR-369-3p was administered one week prior to LPS-induced in order to assess its potential efficacy in ameliorating ALI in mice. The findings indicated that the overexpression of miR-369-3p resulted in enhanced lung function, reduced pulmonary edema, inflammation, and permeability in LPS-induced ALI mice, while the suppression of miR-369-3p exacerbated the damage in these mice. Furthermore, the beneficial effects of TMP on LPS-induced ALI were negated by the downregulation of miR-369-3p. The results of our study demonstrate that TMP mitigates LPS-induced ALI through upregulation of miR-369-3p. Consequently, the findings of this study advocate for the clinical utilization of TMP in ALI treatment, with miR-369-3p emerging as a promising target for future ALI interventions.
Collapse
Affiliation(s)
- Weiting Tao
- Department of Pathophysiology, Bengbu Medical University, Bengbu, Anhui, China
| | - Simin Min
- Suzhou Hospital Affiliated to Anhui Medical University, Suzhou, Anhui, China
| | - Guofeng Chen
- School of Medicine and Health Engineering, Changzhou University, Changzhou, Jiangsu, China
| | - Xu He
- School of Medicine and Health Engineering, Changzhou University, Changzhou, Jiangsu, China
| | - Yuhang Meng
- School of Clinical Medicine, Bengbu Medical University, Bengbu, Anhui, China
| | - Li Li
- Department of Pathophysiology, Bengbu Medical University, Bengbu, Anhui, China
| | - Jie Chen
- Department of Pathophysiology, Bengbu Medical University, Bengbu, Anhui, China
| | - Yan Li
- School of Medicine and Health Engineering, Changzhou University, Changzhou, Jiangsu, China.
| |
Collapse
|
4
|
Gerasimovskaya E, Patil RS, Davies A, Maloney ME, Simon L, Mohamed B, Cherian-Shaw M, Verin AD. Extracellular purines in lung endothelial permeability and pulmonary diseases. Front Physiol 2024; 15:1450673. [PMID: 39234309 PMCID: PMC11372795 DOI: 10.3389/fphys.2024.1450673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 08/05/2024] [Indexed: 09/06/2024] Open
Abstract
The purinergic signaling system is an evolutionarily conserved and critical regulatory circuit that maintains homeostatic balance across various organ systems and cell types by providing compensatory responses to diverse pathologies. Despite cardiovascular diseases taking a leading position in human morbidity and mortality worldwide, pulmonary diseases represent significant health concerns as well. The endothelium of both pulmonary and systemic circulation (bronchial vessels) plays a pivotal role in maintaining lung tissue homeostasis by providing an active barrier and modulating adhesion and infiltration of inflammatory cells. However, investigations into purinergic regulation of lung endothelium have remained limited, despite widespread recognition of the role of extracellular nucleotides and adenosine in hypoxic, inflammatory, and immune responses within the pulmonary microenvironment. In this review, we provide an overview of the basic aspects of purinergic signaling in vascular endothelium and highlight recent studies focusing on pulmonary microvascular endothelial cells and endothelial cells from the pulmonary artery vasa vasorum. Through this compilation of research findings, we aim to shed light on the emerging insights into the purinergic modulation of pulmonary endothelial function and its implications for lung health and disease.
Collapse
Affiliation(s)
| | - Rahul S. Patil
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Adrian Davies
- Department of Internal Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - McKenzie E. Maloney
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Office of Academic Affairs, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Liselle Simon
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Basmah Mohamed
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Mary Cherian-Shaw
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Alexander D. Verin
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
5
|
McGee MY, Ogunsina O, Boshra SN, Gao X, Majetschak M. β-Adrenoceptor Agonists Attenuate Thrombin-Induced Impairment of Human Lung Endothelial Cell Barrier Function and Protect the Lung Vascular Barrier during Resuscitation from Hemorrhagic Shock. Biomedicines 2024; 12:1813. [PMID: 39200278 PMCID: PMC11352179 DOI: 10.3390/biomedicines12081813] [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: 07/10/2024] [Revised: 08/01/2024] [Accepted: 08/07/2024] [Indexed: 09/02/2024] Open
Abstract
β-adrenoceptor (β-AR) agonists are known to antagonize thrombin-induced impairment (TII) of bovine and ovine lung endothelial barrier function. The effects of adrenoceptor agonists and other vasoactive agents on human lung microvascular endothelial cell (HULEC-5a) barrier function upon thrombin exposure have not been studied. Furthermore, it is unknown whether the in vitro effects of adrenoceptor agonists translate to lung protective effects in vivo. We observed that epinephrine, norepinephrine, and phenylephrine enhanced normal and prevented TII of HULEC-5a barrier function. Arginine vasopressin and angiotensin II were ineffective. α1B-, α2A/B-, and β1/2-ARs were detectable in HULEC-5a by RT-PCR. Propranolol but not doxazosin blocked the effects of all adrenoceptor agonists. Phenylephrine stimulated β2-AR-mediated Gαs activation with 13-fold lower potency than epinephrine. The EC50 to inhibit TII of HULEC-5a barrier function was 1.8 ± 1.9 nM for epinephrine and >100 nM for phenylephrine. After hemorrhagic shock and fluid resuscitation in rats, Evans blue extravasation into the lung increased threefold (p < 0.01 vs. sham). Single low-dose (1.8 μg/kg) epinephrine administration at the beginning of resuscitation had no effects on blood pressure and reduced Evans blue extravasation by 60% (p < 0.05 vs. vehicle). Our findings confirm the effects of β-adrenoceptor agonists in HULEC-5a and suggest that low-dose β-adrenoceptor agonist treatment protects lung vascular barrier function after traumatic hemorrhagic shock.
Collapse
Affiliation(s)
- Michelle Y. McGee
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA; (M.Y.M.); (O.O.); (S.N.B.); (X.G.)
| | - Ololade Ogunsina
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA; (M.Y.M.); (O.O.); (S.N.B.); (X.G.)
| | - Sadia N. Boshra
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA; (M.Y.M.); (O.O.); (S.N.B.); (X.G.)
- Department of Chemistry, University of South Florida, Tampa, FL 33612, USA
| | - Xianlong Gao
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA; (M.Y.M.); (O.O.); (S.N.B.); (X.G.)
| | - Matthias Majetschak
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA; (M.Y.M.); (O.O.); (S.N.B.); (X.G.)
- Department of Molecular Pharmacology & Physiology, University of South Florida, Tampa, FL 33612, USA
| |
Collapse
|
6
|
Yang B, Hang S, Xu S, Gao Y, Yu W, Zang G, Zhang L, Wang Z. Macrophage polarisation and inflammatory mechanisms in atherosclerosis: Implications for prevention and treatment. Heliyon 2024; 10:e32073. [PMID: 38873669 PMCID: PMC11170185 DOI: 10.1016/j.heliyon.2024.e32073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 05/11/2024] [Accepted: 05/28/2024] [Indexed: 06/15/2024] Open
Abstract
Atherosclerosis is a chronic inflammatory disease characterised by plaque accumulation in the arteries. Macrophages are immune cells that are crucial in the development of atherosclerosis. Macrophages can adopt different phenotypes, with the M1 phenotype promoting inflammation while the M2 phenotype counteracting it. This review focuses on the factors that drive the polarisation of M1 macrophages towards a pro-inflammatory phenotype during AS. Additionally, we explored metabolic reprogramming mechanisms and cytokines secretion by M1 macrophages. Hyperlipidaemia is widely recognised as a major risk factor for atherosclerosis. Modified lipoproteins released in the presence of hyperlipidaemia can trigger the release of cytokines and recruit circulating monocytes, which adhere to the damaged endothelium and differentiate into macrophages. Macrophages engulf lipids, leading to the formation of foam cells. As atherosclerosis progresses, foam cells become the necrotic core within the atherosclerotic plaques, destabilising them and triggering ischaemic disease. Furthermore, we discuss recent research focusing on targeting macrophages or inflammatory pathways for preventive or therapeutic purposes. These include statins, PCSK9 inhibitors, and promising nanotargeted drugs. These new developments hold the potential for the prevention and treatment of atherosclerosis and its related complications.
Collapse
Affiliation(s)
- Bo Yang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Sanhua Hang
- Department of Hematology, Affiliated Danyang Hospital of Nantong University, Danyang, 212300, China
| | - Siting Xu
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Yun Gao
- Department of Pathology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Wenhua Yu
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Guangyao Zang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Lili Zhang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| |
Collapse
|
7
|
Bavuso M, Miller N, Sill JM, Dobrian A, Colunga Biancatelli RML. Extracellular vesicles in acute respiratory distress syndrome: Understanding protective and harmful signaling for the development of new therapeutics. Histol Histopathol 2024; 39:131-144. [PMID: 37712224 DOI: 10.14670/hh-18-659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/16/2023]
Abstract
Acute respiratory distress syndrome (ARDS) is a severe respiratory condition characterized by increased lung permeability, hyper-inflammatory state, and fluid leak into the alveolar spaces. ARDS is a heterogeneous disease, with multiple direct and indirect causes that result in a mortality of up to 40%. Due to the ongoing Covid-19 pandemic, its incidence has increased up to ten-fold. Extracellular vesicles (EVs) are small liposome-like particles that mediate intercellular communication and play a major role in ARDS pathophysiology. Indeed, they participate in endothelial barrier dysfunction and permeability, neutrophil, and macrophage activation, and also in the development of a hypercoagulable state. A more thorough understanding of the variegated and cell-specific functions of EVs may lead to the development of safe and effective therapeutics. In this review, we have collected evidence of EVs role in ARDS, revise the main mechanisms of production and internalization and summarize the current therapeutical approaches that have shown the ability to modulate EV signaling.
Collapse
Affiliation(s)
- Matthew Bavuso
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Noel Miller
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Joshua M Sill
- Division of Pulmonary and Critical Care, Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, Virginia, USA
| | - Anca Dobrian
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Ruben M L Colunga Biancatelli
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA, USA
- Division of Pulmonary and Critical Care, Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, Virginia, USA
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA, USA.
| |
Collapse
|
8
|
Mahamed Z, Shadab M, Najar RA, Millar MW, Bal J, Pressley T, Fazal F. The Protective Role of Mitochondria-Associated Endoplasmic Reticulum Membrane (MAM) Protein Sigma-1 Receptor in Regulating Endothelial Inflammation and Permeability Associated with Acute Lung Injury. Cells 2023; 13:5. [PMID: 38201208 PMCID: PMC10778450 DOI: 10.3390/cells13010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/01/2023] [Accepted: 12/12/2023] [Indexed: 01/12/2024] Open
Abstract
Earlier studies from our lab identified endoplasmic reticulum (ER) chaperone BiP/GRP78, an important component of MAM, to be a novel determinant of endothelial cell (EC) dysfunction associated with acute lung injury (ALI). Sigma1R (Sig1R) is another unique ER receptor chaperone that has been identified to associate with BiP/GRP78 at the MAM and is known to be a pluripotent modulator of cellular homeostasis. However, it is unclear if Sig1R also plays a role in regulating the EC inflammation and permeability associated with ALI. Our data using human pulmonary artery endothelial cells (HPAECs) showed that siRNA-mediated knockdown of Sig1R potentiated LPS-induced the expression of proinflammatory molecules ICAM-1, VCAM-1 and IL-8. Consistent with this, Sig1R agonist, PRE-084, known to activate Sig1R by inducing its dissociation from BiP/GRP78, blunted the above response. Notably, PRE-084 failed to blunt LPS-induced inflammatory responses in Sig1R-depleted cells, confirming that the effect of PRE-084 is driven by Sig1R. Furthermore, Sig1R antagonist, NE-100, known to inactivate Sig1R by blocking its dissociation from BiP/GRP78, failed to block LPS-induced inflammatory responses, establishing that dissociation from BiP/GRP78 is required for Sig1R to exert its anti-inflammatory action. Unlike Sig1R, the siRNA-mediated knockdown or Subtilase AB-mediated inactivation of BiP/GRP78 protected against LPS-induced EC inflammation. Interestingly, the protective effect of BiP/GRP78 knockdown or inactivation was abolished in cells that were depleted of Sig1R, confirming that BiP/GRP78 knockdown/inactivation-mediated suppression of EC inflammation is mediated via Sig1R. In view of these findings, we determined the in vivo relevance of Sig1R in a mouse model of sepsis-induced ALI. The intraperitoneal injection of PRE-084 mitigated sepsis-induced ALI, as evidenced by a decrease in ICAM-1, IL-6 levels, lung PMN infiltration, and lung vascular leakage. Together, these data evidence a protective role of Sig1R against endothelial dysfunction associated with ALI and identify it as a viable target in terms of controlling ALI in sepsis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Fabeha Fazal
- Department of Pediatrics (Neonatology), Lung Biology and Disease Program, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA; (Z.M.); (M.S.); (R.A.N.); (M.W.M.); (J.B.); (T.P.)
| |
Collapse
|
9
|
Chen X, Chen J, Liu S, Li X. Everolimus-induced hyperpermeability of endothelial cells causes lung injury. Exp Biol Med (Maywood) 2023; 248:2440-2448. [PMID: 38158699 PMCID: PMC10903245 DOI: 10.1177/15353702231220672] [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: 06/20/2023] [Accepted: 10/06/2023] [Indexed: 01/03/2024] Open
Abstract
The mammalian target of rapamycin (mTOR) inhibitors, everolimus (but not dactolisib), is frequently associated with lung injury in clinical therapies. However, the underlying mechanisms remain unclear. Endothelial cell barrier dysfunction plays a major role in the pathogenesis of the lung injury. This study hypothesizes that everolimus increases pulmonary endothelial permeability, which leads to lung injury. We tested the effects of everolimus on human pulmonary microvascular endothelial cell (HPMEC) permeability and a mouse model of intraperitoneal injection of everolimus was established to investigate the effect of everolimus on pulmonary vascular permeability. Our data showed that everolimus increased human pulmonary microvascular endothelial cell (HPMEC) permeability which was associated with MLC phosphorylation and F-actin stress fiber formation. Furthermore, everolimus induced an increasing concentration of intracellular calcium Ca2+ leakage in HPMECs and this was normalized with ryanodine pretreatment. In addition, ryanodine decreased everolimus-induced phosphorylation of PKCα and MLC, and barrier disruption in HPMECs. Consistent with in vitro data, everolimus treatment caused a visible lung-vascular barrier dysfunction, including an increase in protein in BALF and lung capillary-endothelial permeability, which was significantly attenuated by pretreatment with an inhibitor of PKCα, MLCK, and ryanodine. This study shows that everolimus induced pulmonary endothelial hyper-permeability, at least partly, in an MLC phosphorylation-mediated EC contraction which is influenced in a Ca2+-dependent manner and can lead to lung injury through mTOR-independent mechanisms.
Collapse
Affiliation(s)
- Xiaolin Chen
- Department of Clinical Laboratory, Pingxiang People's Hospital, Pingxiang 337000, China
- Department of Clinical Laboratory, The Sixth Clinical College of Gannan Medical University, Pingxiang 337000, China
| | - Jianhui Chen
- Department of Clinical Laboratory, Pingxiang People's Hospital, Pingxiang 337000, China
| | - Shuihong Liu
- Department of Clinical Laboratory, Pingxiang People's Hospital, Pingxiang 337000, China
| | - Xianfan Li
- Department of Clinical Laboratory, Pingxiang People's Hospital, Pingxiang 337000, China
| |
Collapse
|
10
|
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
|
11
|
Choroszy M, Środa-Pomianek K, Wawrzyńska M, Chmielarz M, Bożemska E, Sobieszczańska B. The Role of Palmitic Acid in the Co-Toxicity of Bacterial Metabolites to Endothelial Cells. Vasc Health Risk Manag 2023; 19:399-409. [PMID: 37426328 PMCID: PMC10329449 DOI: 10.2147/vhrm.s408897] [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: 02/17/2023] [Accepted: 05/18/2023] [Indexed: 07/11/2023] Open
Abstract
Introduction Metabolic endotoxemia most often results from obesity and is accompanied by an increase in the permeability of the intestinal epithelial barrier, allowing co-absorption of bacterial metabolites and diet-derived fatty acids into the bloodstream. A high-fat diet (HFD) leading to obesity is a significant extrinsic factor in developing vascular atherosclerosis. In this study, we evaluated the effects of palmitic acid (PA) as a representative of long-chain saturated fatty acids (LCSFA) commonly present in HFDs, along with endotoxin (LPS; lipopolysaccharide) and uremic toxin indoxyl sulfate (IS), on human vascular endothelial cells (HUVECs). Methods HUVECs viability was measured based on tetrazolium salt metabolism, and cell morphology was assessed with fluorescein-phalloidin staining of cells' actin cytoskeleton. The effects of simultaneous treatment of endothelial cells with PA, LPS, and IS on nitro-oxidative stress in vascular cells were evaluated quantitatively with fluorescent probes. The expression of vascular cell adhesion molecule VCAM-1, E-selectin, and occludin, an essential tight junction protein, in HUVECs treated with these metabolites was evaluated in Western blot. Results PA, combined with LPS and IS, did not influence HUVECs viability but induced stress on actin fibers and focal adhesion complexes. Moreover, PA combined with LPS significantly enhanced reactive oxygen species (ROS) production in HUVECs but decreased nitric oxide (NO) generation. PA also considerably increased the expression of VCAM-1 and E-selectin in HUVECs treated with LPS or IS but decreased occludin expression. Conclusion Palmitic acid enhances the toxic effect of metabolic endotoxemia on the vascular endothelium.
Collapse
Affiliation(s)
- Marcin Choroszy
- Department of Microbiology, Wroclaw Medical University, Wroclaw, Poland
| | - Kamila Środa-Pomianek
- Department of Biophysics and Neuroscience, Wroclaw Medical University, Wroclaw, Poland
| | - Magdalena Wawrzyńska
- Department of Preclinical Studies, Faculty of Health Sciences, Wroclaw Medical University, Wroclaw, Poland
| | - Mateusz Chmielarz
- Department of Microbiology, Wroclaw Medical University, Wroclaw, Poland
| | - Edyta Bożemska
- Department of Microbiology, Wroclaw Medical University, Wroclaw, Poland
| | | |
Collapse
|
12
|
Siddiqui MR, Reddy NM, Faridi HM, Shahid M, Shanley TP. Metformin alleviates lung-endothelial hyperpermeability by regulating cofilin-1/PP2AC pathway. Front Pharmacol 2023; 14:1211460. [PMID: 37361221 PMCID: PMC10285707 DOI: 10.3389/fphar.2023.1211460] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 05/22/2023] [Indexed: 06/28/2023] Open
Abstract
Background: Microvascular endothelial hyperpermeability is an earliest pathological hallmark in Acute Lung Injury (ALI), which progressively leads to Acute Respiratory Distress Syndrome (ARDS). Recently, vascular protective and anti-inflammatory effect of metformin, irrespective of glycemic control, has garnered significant interest. However, the underlying molecular mechanism(s) of metformin's barrier protective benefits in lung-endothelial cells (ECs) has not been clearly elucidated. Many vascular permeability-increasing agents weakened adherens junctions (AJ) integrity by inducing the reorganization of the actin cytoskeleton and stress fibers formation. Here, we hypothesized that metformin abrogated endothelial hyperpermeability and strengthen AJ integrity via inhibiting stress fibers formation through cofilin-1-PP2AC pathway. Methods: We pretreated human lung microvascular ECs (human-lung-ECs) with metformin and then challenged with thrombin. To investigate the vascular protective effects of metformin, we studied changes in ECs barrier function using electric cell-substrate impedance sensing, levels of actin stress fibers formation and inflammatory cytokines IL-1β and IL-6 expression. To explore the downstream mechanism, we studied the Ser3-phosphorylation-cofilin-1 levels in scramble and PP2AC-siRNA depleted ECs in response to thrombin with and without metformin pretreatment. Results: In-vitro analyses showed that metformin pretreatment attenuated thrombin-induced hyperpermeability, stress fibers formation, and the levels of inflammatory cytokines IL-6 and IL-β in human-lung-ECs. We found that metformin mitigated Ser3-phosphorylation mediated inhibition of cofilin-1 in response to thrombin. Furthermore, genetic deletion of PP2AC subunit significantly inhibited metformin efficacy to mitigate thrombin-induced Ser3-phosphorylation cofilin-1, AJ disruption and stress fibers formation. We further demonstrated that metformin increases PP2AC activity by upregulating PP2AC-Leu309 methylation in human-lung-ECs. We also found that the ectopic expression of PP2AC dampened thrombin-induced Ser3-phosphorylation-mediated inhibition of cofilin-1, stress fibers formation and endothelial hyperpermeability. Conclusion: Together, these data reveal the unprecedented endothelial cofilin-1/PP2AC signaling axis downstream of metformin in protecting against lung vascular endothelial injury and inflammation. Therefore, pharmacologically enhancing endothelial PP2AC activity may lead to the development of novel therapeutic approaches for prevention of deleterious effects of ALI on vascular ECs.
Collapse
Affiliation(s)
- M. Rizwan Siddiqui
- Department of Pediatrics, Ann & Robert H. Lurie Children’s Hospital of Chicago, Stanley Manne Children’s Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Narsa M. Reddy
- Department of Pediatrics, Ann & Robert H. Lurie Children’s Hospital of Chicago, Stanley Manne Children’s Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Hafeez M. Faridi
- Drug Discovery Center, Department of Internal Medicine, Rush University Medical Center, Chicago, IL, United States
| | - Mohd Shahid
- Department of Pharmaceutical Sciences, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Thomas P. Shanley
- Department of Pediatrics, Ann & Robert H. Lurie Children’s Hospital of Chicago, Stanley Manne Children’s Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
13
|
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
|
14
|
Luo G, Liu B, Fu T, Liu Y, Li B, Li N, Geng Q. The Role of Histone Deacetylases in Acute Lung Injury-Friend or Foe. Int J Mol Sci 2023; 24:ijms24097876. [PMID: 37175583 PMCID: PMC10178380 DOI: 10.3390/ijms24097876] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/04/2023] [Accepted: 04/14/2023] [Indexed: 05/15/2023] Open
Abstract
Acute lung injury (ALI), caused by intrapulmonary or extrapulmonary factors such as pneumonia, shock, and sepsis, eventually disrupts the alveolar-capillary barrier, resulting in diffuse pulmonary oedema and microatasis, manifested by refractory hypoxemia, and respiratory distress. Not only is ALI highly lethal, but even if a patient survives, there are also multiple sequelae. Currently, there is no better treatment than supportive care, and we urgently need to find new targets to improve ALI. Histone deacetylases (HDACs) are epigenetically important enzymes that, together with histone acetylases (HATs), regulate the acetylation levels of histones and non-histones. While HDAC inhibitors (HDACis) play a therapeutic role in cancer, inflammatory, and neurodegenerative diseases, there is also a large body of evidence suggesting the potential of HDACs as therapeutic targets in ALI. This review explores the unique mechanisms of HDACs in different cell types of ALI, including macrophages, pulmonary vascular endothelial cells (VECs), alveolar epithelial cells (AECs), and neutrophils.
Collapse
Affiliation(s)
- Guoqing Luo
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Bohao Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Tinglv Fu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yi Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Boyang Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
15
|
Liu J, Rickel A, Smith S, Hong Z, Wang C. "Non-cytotoxic" doses of metal-organic framework nanoparticles increase endothelial permeability by inducing actin reorganization. J Colloid Interface Sci 2023; 634:323-335. [PMID: 36535168 PMCID: PMC9840705 DOI: 10.1016/j.jcis.2022.12.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/28/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022]
Abstract
Cytotoxicity of nanoparticles is routinely characterized by biochemical assays such as cell viability and membrane integrity assays. However, these approaches overlook cellular biophysical properties including changes in the actin cytoskeleton, cell stiffness, and cell morphology, particularly when cells are exposed to "non-cytotoxic" doses of nanoparticles. Zeolitic imidazolate framework-8 nanoparticles (ZIF-8 NPs), a member of metal-organic framework family, has received increasing interest in various fields such as environmental and biomedical sciences. ZIF-8 NPs may enter the blood circulation system after unintended oral and inhalational exposure or intended intravenous injection for diagnostic and therapeutic applications, yet the effect of ZIF-8 NPs on vascular endothelial cells is not well understood. Here, the biophysical impact of "non-cytotoxic" dose ZIF-8 NPs on human aortic endothelial cells (HAECs) is investigated. We demonstrate that "non-cytotoxic" doses of ZIF-8 NPs, pre-defined by a series of biochemical assays, can increase the endothelial permeability of HAEC monolayers by causing cell junction disruption and intercellular gap formation, which can be attributed to actin reorganization within adjacent HAECs. Nanomechanical atomic force microscopy and super resolution fluorescence microscopy further confirm that "non-cytotoxic" doses of ZIF-8 NPs change the actin structure and cell morphology of HAECs at the single cell level. Finally, the underlying mechanism of actin reorganization induced by the "non-cytotoxic" dose ZIF-8 NPs is elucidated. Together, this study indicates that the "non-cytotoxic" doses of ZIF-8 NPs, intentionally or unintentionally introduced into blood circulation, may still pose a threat to human health, considering increased endothelial permeability is essential to the progression of a variety of diseases. From a broad view of cytotoxicity evaluation, it is important to consider the biophysical properties of cells, since they can serve as novel and more sensitive markers to assess nanomaterial's cytotoxicity.
Collapse
Affiliation(s)
- Jinyuan Liu
- Nanoscience and Nanoengineering, South Dakota School of Mines and Technology, 501 East Saint Joseph Street, Rapid City, SD 57701, USA; BioSystems, Networks & Translational Research (BioSNTR), 501 East Saint Joseph Street, Rapid City, SD 57701, USA
| | - Alex Rickel
- Biomedical Engineering, University of South Dakota, 4800 N Career Avenue, Sioux Falls, SD 57107, USA; BioSystems, Networks & Translational Research (BioSNTR), 501 East Saint Joseph Street, Rapid City, SD 57701, USA
| | - Steve Smith
- Nanoscience and Nanoengineering, South Dakota School of Mines and Technology, 501 East Saint Joseph Street, Rapid City, SD 57701, USA; BioSystems, Networks & Translational Research (BioSNTR), 501 East Saint Joseph Street, Rapid City, SD 57701, USA
| | - Zhongkui Hong
- Biomedical Engineering, University of South Dakota, 4800 N Career Avenue, Sioux Falls, SD 57107, USA; BioSystems, Networks & Translational Research (BioSNTR), 501 East Saint Joseph Street, Rapid City, SD 57701, USA; Mechanical Engineering, Texas Tech University, 805 Boston Ave, Lubbock, TX 79409, USA.
| | - Congzhou Wang
- Nanoscience and Nanoengineering, South Dakota School of Mines and Technology, 501 East Saint Joseph Street, Rapid City, SD 57701, USA; BioSystems, Networks & Translational Research (BioSNTR), 501 East Saint Joseph Street, Rapid City, SD 57701, USA.
| |
Collapse
|
16
|
Significance of Pulmonary Endothelial Injury and the Role of Cyclooxygenase-2 and Prostanoid Signaling. BIOENGINEERING (BASEL, SWITZERLAND) 2023; 10:bioengineering10010117. [PMID: 36671689 PMCID: PMC9855370 DOI: 10.3390/bioengineering10010117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/19/2023]
Abstract
The endothelium plays a key role in the dynamic balance of hemodynamic, humoral and inflammatory processes in the human body. Its central importance and the resulting therapeutic concepts are the subject of ongoing research efforts and form the basis for the treatment of numerous diseases. The pulmonary endothelium is an essential component for the gas exchange in humans. Pulmonary endothelial dysfunction has serious consequences for the oxygenation and the gas exchange in humans with the potential of consecutive multiple organ failure. Therefore, in this review, the dysfunction of the pulmonary endothel due to viral, bacterial, and fungal infections, ventilator-related injury, and aspiration is presented in a medical context. Selected aspects of the interaction of endothelial cells with primarily alveolar macrophages are reviewed in more detail. Elucidation of underlying causes and mechanisms of damage and repair may lead to new therapeutic approaches. Specific emphasis is placed on the processes leading to the induction of cyclooxygenase-2 and downstream prostanoid-based signaling pathways associated with this enzyme.
Collapse
|
17
|
Li J, Zhang M, He Y, Du YH, Zhang XZ, Georgi R, Kolberg B, Xu YL. Molecular Mechanism of Electroacupuncture Regulating Cerebral Arterial Contractile Protein in Rats with Cerebral Infarction Based on MLCK Pathway. Chin J Integr Med 2023; 29:61-68. [PMID: 35344120 DOI: 10.1007/s11655-022-3468-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/08/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To explore the effect of electroacupuncture (EA) intervention on the vasoconstriction of cerebral artery smooth muscle cells after cerebral infarction. METHODS Male Wistar rats were randomly divided into 3 groups by a random number table: the model group (n=24), the EA group (n=24), and the normal group (n=6). The model and the EA groups were divided into different time subgroups at 0.5, 1, 3, and 6 h after middle cerebral artery occlusion (MCAO), with 6 rats in each subgroup. MCAO model was established using intraluminal suture occlusion method. The EA group was given EA treatment at acupoint Shuigou (GV 26) instantly after MCAO for 20 min. The contents of cerebrovascular smooth muscle MLCK, the 3 subunits of myosin light chain phosphatase (MLCP) MYPT1, PP1c-δ and M20, as well as myosin-ATPase activity were detected using immunohistochemistry and Western blotting. RESULTS The overall expression level of the MYPT1 and PP1c-δ in the model group was significantly higher (P<0.01). After EA intervention, the 0.5 h group expression level was close to that of the normal group (P>0.05), and the other subgroups were still significantly higher than the normal group (P<0.01). After EA intervention, the expression level of each subgroup was significantly lower than the corresponding model group. There was a significant difference between the 0.5 and 1 h subgroups (P<0.01), while a difference was also observed between the 3 and 6 h subgroups (P<0.05). The dynamic change rule gradually increased with the prolongation of infarction time within 6 h after infarction. CONCLUSION EA intervention can inhibit contraction of cerebral vascular smooth muscle cells and regulate smooth muscle relaxation by regulating MLCK pathway.
Collapse
Affiliation(s)
- Jing Li
- Institute of Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
| | - Min Zhang
- Institute of Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
| | - Ying He
- Institute of Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
| | - Yuan-Hao Du
- Institute of Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China.
| | - Xue-Zhu Zhang
- Institute of Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
| | - Rainer Georgi
- Max Planck Institute for Medical Research, Heidelberg, 69120, Germany
| | | | - Yan-Long Xu
- Department of Acupuncture and Moxibustion, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, 730050, China
| |
Collapse
|
18
|
Ramasubramanian B, Kim J, Ke Y, Li Y, Zhang CO, Promnares K, Tanaka KA, Birukov KG, Karki P, Birukova AA. Mechanisms of pulmonary endothelial permeability and inflammation caused by extracellular histone subunits H3 and H4. FASEB J 2022; 36:e22470. [PMID: 35969180 DOI: 10.1096/fj.202200303rr] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 07/04/2022] [Accepted: 07/14/2022] [Indexed: 11/11/2022]
Abstract
Extracellular DNA-binding proteins such as histones are danger-associated molecular pattern released by the injured tissues in trauma and sepsis settings, which trigger host immune response and vascular dysfunction. Molecular events leading to histone-induced endothelial cell (EC) dysfunction remain poorly understood. This study performed comparative analysis of H1, H2A, H2B, H3, and H4 histone subunits effects on human pulmonary EC permeability and inflammatory response. Analysis of transendothelial electrical resistance and EC monolayer permeability for macromolecues revealed that H3 and H4, but not H1, H2A, or H2B caused dose-dependent EC permeability accompanied by disassembly of adherens junctions. At higher doses, H3 and H4 activated nuclear factor kappa B inflammatory cascade leading to upregulation EC adhesion molecules ICAM1, VCAM1, E-selectin, and release of inflammatory cytokines. Inhibitory receptor analysis showed that toll-like receptor (TLR) 4 but not TLR1/2 or receptor for advanced glycation end inhibition significantly attenuated deleterious effects of H3 and H4 histones. Inhibitor of Rho-kinase was without effect, while inhibition of Src kinase caused partial preservation of cell-cell junctions, H3/H4-induced permeability and inflammation. Deleterious effects of H3/H4 were blocked by heparin. Activation of Epac-Rap1 signaling restored EC barrier properties after histone challenge. Intravenous injection of histones in mice caused elevation of inflammatory markers and increased vascular leak. Post-treatment with pharmacological Epac/Rap1 activator suppressed injurious effects of histones in vitro and in vivo. These results identify H3 and H4 as key histone subunits exhibiting deleterious effects on pulmonary vascular endothelium via TLR4-dependent mechanism. In conclusion, elevation of circulating histones may represent a serious risk of exacerbated acute lung injury (ALI) and multiple organ injury during severe trauma and infection.
Collapse
Affiliation(s)
- Baalachandran Ramasubramanian
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Junghyun Kim
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Yunbo Ke
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Yue Li
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Chen-Ou Zhang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Kamoltip Promnares
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Kenichi A Tanaka
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Konstantin G Birukov
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Pratap Karki
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Anna A Birukova
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
19
|
Parinandi N, Gerasimovskaya E, Verin A. Editorial: Molecular mechanisms of lung endothelial permeability. Front Physiol 2022; 13:976873. [PMID: 35936898 PMCID: PMC9355505 DOI: 10.3389/fphys.2022.976873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 06/30/2022] [Indexed: 01/16/2023] Open
Affiliation(s)
- Narasimham Parinandi
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Evgenia Gerasimovskaya
- Division of Critical Care Medicine, Department of Pediatrics, University of Colorado Denver, Aurora, CO, United States
| | - Alexander Verin
- Vascular Biology Center and Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, United States,*Correspondence: Alexander Verin,
| |
Collapse
|
20
|
Role of ARP2/3 Complex-Driven Actin Polymerization in RSV Infection. Pathogens 2021; 11:pathogens11010026. [PMID: 35055974 PMCID: PMC8781601 DOI: 10.3390/pathogens11010026] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/14/2021] [Accepted: 12/20/2021] [Indexed: 01/30/2023] Open
Abstract
Respiratory syncytial virus (RSV) is the leading viral agent causing bronchiolitis and pneumonia in children under five years old worldwide. The RSV infection cycle starts with macropinocytosis-based entry into the host airway epithelial cell membrane, followed by virus transcription, replication, assembly, budding, and spread. It is not surprising that the host actin cytoskeleton contributes to different stages of the RSV replication cycle. RSV modulates actin-related protein 2/3 (ARP2/3) complex-driven actin polymerization for a robust filopodia induction on the infected lung epithelial A549 cells, which contributes to the virus’s budding, and cell-to-cell spread. Thus, a comprehensive understanding of RSV-induced cytoskeletal modulation and its role in lung pathobiology may identify novel intervention strategies. This review will focus on the role of the ARP2/3 complex in RSV’s pathogenesis and possible therapeutic targets to the ARP2/3 complex for RSV.
Collapse
|
21
|
D’Agnillo F, Walters KA, Xiao Y, Sheng ZM, Scherler K, Park J, Gygli S, Rosas LA, Sadtler K, Kalish H, Blatti CA, Zhu R, Gatzke L, Bushell C, Memoli MJ, O’Day SJ, Fischer TD, Hammond TC, Lee RC, Cash JC, Powers ME, O’Keefe GE, Butnor KJ, Rapkiewicz AV, Travis WD, Layne SP, Kash JC, Taubenberger JK. Lung epithelial and endothelial damage, loss of tissue repair, inhibition of fibrinolysis, and cellular senescence in fatal COVID-19. Sci Transl Med 2021; 13:eabj7790. [PMID: 34648357 PMCID: PMC11000440 DOI: 10.1126/scitranslmed.abj7790] [Citation(s) in RCA: 140] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is characterized by respiratory distress, multiorgan dysfunction, and, in some cases, death. The pathological mechanisms underlying COVID-19 respiratory distress and the interplay with aggravating risk factors have not been fully defined. Lung autopsy samples from 18 patients with fatal COVID-19, with symptom onset-to-death times ranging from 3 to 47 days, and antemortem plasma samples from 6 of these cases were evaluated using deep sequencing of SARS-CoV-2 RNA, multiplex plasma protein measurements, and pulmonary gene expression and imaging analyses. Prominent histopathological features in this case series included progressive diffuse alveolar damage with excessive thrombosis and late-onset pulmonary tissue and vascular remodeling. Acute damage at the alveolar-capillary barrier was characterized by the loss of surfactant protein expression with injury to alveolar epithelial cells, endothelial cells, respiratory epithelial basal cells, and defective tissue repair processes. Other key findings included impaired clot fibrinolysis with increased concentrations of plasma and lung plasminogen activator inhibitor-1 and modulation of cellular senescence markers, including p21 and sirtuin-1, in both lung epithelial and endothelial cells. Together, these findings further define the molecular pathological features underlying the pulmonary response to SARS-CoV-2 infection and provide important insights into signaling pathways that may be amenable to therapeutic intervention.
Collapse
Affiliation(s)
- Felice D’Agnillo
- Laboratory of Biochemistry and Vascular Biology, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | | | - Yongli Xiao
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Zong-Mei Sheng
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | - Jaekeun Park
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sebastian Gygli
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Luz Angela Rosas
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kaitlyn Sadtler
- Section on Immunoengineering, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Heather Kalish
- Bioengineering and Physical Sciences Shared Resource, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Charles A. Blatti
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Ruoqing Zhu
- Department of Statistics, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Lisa Gatzke
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Colleen Bushell
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Matthew J. Memoli
- Clinical Studies Unit, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | - Raymond C. Lee
- Division of Cardiothoracic Surgery, USC Keck School of Medicine, Los Angeles, CA, USA
| | - J. Christian Cash
- Division of Cardiothoracic Surgery, USC Keck School of Medicine, Los Angeles, CA, USA
| | - Matthew E. Powers
- Division of Cardiothoracic Surgery, USC Keck School of Medicine, Los Angeles, CA, USA
| | - Grant E. O’Keefe
- Department of Surgery, University of Washington, Harborview Medical Center, Seattle, WA, USA
| | - Kelly J. Butnor
- Department of Pathology and Laboratory Medicine, University of Vermont Medical Center, Burlington, VT, USA
| | - Amy V. Rapkiewicz
- Department of Pathology, New York University Long Island School of Medicine, Mineola, NY, USA
| | - William D. Travis
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - John C. Kash
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jeffery K. Taubenberger
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
22
|
Karki P, Birukova AA. Microtubules as Major Regulators of Endothelial Function: Implication for Lung Injury. Front Physiol 2021; 12:758313. [PMID: 34777018 PMCID: PMC8582326 DOI: 10.3389/fphys.2021.758313] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 10/06/2021] [Indexed: 01/04/2023] Open
Abstract
Endothelial dysfunction has been attributed as one of the major complications in COVID-19 patients, a global pandemic that has already caused over 4 million deaths worldwide. The dysfunction of endothelial barrier is characterized by an increase in endothelial permeability and inflammatory responses, and has even broader implications in the pathogenesis of acute respiratory syndromes such as ARDS, sepsis and chronic illnesses represented by pulmonary arterial hypertension and interstitial lung disease. The structural integrity of endothelial barrier is maintained by cytoskeleton elements, cell-substrate focal adhesion and adhesive cell junctions. Agonist-mediated changes in endothelial permeability are directly associated with reorganization of actomyosin cytoskeleton leading to cell contraction and opening of intercellular gaps or enhancement of cortical actin cytoskeleton associated with strengthening of endothelial barrier. The role of actin cytoskeleton remodeling in endothelial barrier regulation has taken the central stage, but the impact of microtubules in this process remains less explored and under-appreciated. This review will summarize the current knowledge on the crosstalk between microtubules dynamics and actin cytoskeleton remodeling, describe the signaling mechanisms mediating this crosstalk, discuss epigenetic regulation of microtubules stability and its nexus with endothelial barrier maintenance, and overview a role of microtubules in targeted delivery of signaling molecules regulating endothelial permeability and inflammation.
Collapse
Affiliation(s)
- Pratap Karki
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Anna A Birukova
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
23
|
Altered Moesin and Actin Cytoskeleton Protein Rearrangements Affect Transendothelial Permeability in Human Endothelial Cells upon Dengue Virus Infection and TNF-α Treatment. Viruses 2021; 13:v13102042. [PMID: 34696472 PMCID: PMC8537470 DOI: 10.3390/v13102042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 12/12/2022] Open
Abstract
It has been hypothesized that the host, viral factors, and secreted cytokines (especially TNF-α) play roles in the pathogenesis of secondary dengue infections. Mass spectrometry-based proteomic screening of cytoskeleton fractions isolated from human endothelial (EA.hy926) cells upon dengue virus (DENV) infection and TNF-α treatment identified 450 differentially altered proteins. Among them, decreased levels of moesin, actin stress fiber rearrangements, and dot-like formations of vinculin were observed with western blot analyses and/or immunofluorescence staining (IFA). In vitro vascular permeability assays using EA.hy926 cells, seeded on collagen-coated transwell inserts, showed low levels of transendothelial electrical resistance in treated cells. The synergistic effects of DENV infection and TNF-α treatment caused cellular permeability changes in EA.hy926 cells, which coincided with decreasing moesin levels and the production of abnormal organizations of actin stress fibers and vinculin. Functional studies demonstrated moesin overexpression restored transendothelial permeability in DENV/TNF-α-treated EA.hy926 cells. The present study improves the understanding of the disruption mechanisms of cytoskeleton proteins in enhancing vascular permeability during DENV infection and TNF-α treatment. The study also suggests that these disruption mechanisms are major factors contributing to vascular leakage in severe dengue patients.
Collapse
|
24
|
Shirvaliloo M. The blood-gas barrier in COVID-19: an overview of the effects of SARS-CoV-2 infection on the alveolar epithelial and endothelial cells of the lung. Tissue Barriers 2021; 9:1937013. [PMID: 34232823 PMCID: PMC8794501 DOI: 10.1080/21688370.2021.1937013] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/21/2021] [Accepted: 05/25/2021] [Indexed: 02/08/2023] Open
Abstract
Blood-gas barrier (BGB) or alveolar-capillary barrier is the primary tissue barrier affected by coronavirus disease 2019 (COVID-19). Comprising alveolar epithelial cells (AECs), endothelial cells (ECs) and the extracellular matrix (ECM) in between, the BGB is damaged following the action of multiple pro-inflammatory cytokines during acute inflammation. The infection of AECs and ECs with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the pathogen behind COVID-19, triggers an inflammatory response at the BGB, inducing the release of interleukin 1 (IL-1), IL-6, tumor necrosis factor alpha (TNF-α), transforming growth factor beta (TGF-β), high mobility group box 1 (HMGB1), matrix metalloproteinases (MMPs), intercellular adhesion molecule-1 (ICAM-1) and platelet activating factor (PAF). The end result is the disassembly of adherens junctions (AJs) and tight junctions (TJs) in both AECs and ECs, AEC hyperplasia, EC pyroptosis, ECM remodeling and deposition of fibrin clots in the alveolar capillaries, leading to disintegration and thickening of the BGB, and ultimately, hypoxia. This commentary seeks to provide a brief account of how the BGB might become affected in COVID-19.
Collapse
Affiliation(s)
- Milad Shirvaliloo
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
25
|
Zhang N, Wang G, Sun G. Actin-binding protein, IQGAP1, regulates LPS-induced RPMVECs hyperpermeability and ICAM-1 upregulation via Rap1/Src signalling pathway. Cell Signal 2021; 85:110067. [PMID: 34147590 DOI: 10.1016/j.cellsig.2021.110067] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/11/2021] [Accepted: 06/15/2021] [Indexed: 11/30/2022]
Abstract
Pulmonary microvascular barrier dysfunction is a hallmark feature of acute lung injury (ALI). IQGAP1 is a ubiquitously expressed scaffolding protein known to regulate cancer metastasis, angiogenesis, and barrier stability. However, the function of IQGAP1 in lipopolysaccharide (LPS)-induced microvascular endothelial hyperpermeability remains poorly understood. In the present study, we demonstrated that IQGAP1 was markedly upregulated in LPS-induced ALI models and rat pulmonary microvascular endothelial cells (RPMVECs). Lentivirus-mediated knockdown of IQGAP1 significantly attenuated the formation of actin stress fibers, phosphorylation of myosin light chain (MLC), and disruption of VE-cadherin, thereby protecting the RPMVECs barrier failure from LPS damage. In addition, IQGAP1 depletion reduced the reactive oxygen species (ROS)-mediated increase in intracellular adhesion molecule-1 (ICAM-1) in RPMVECs stimulated with LPS. Mechanistically, we found that the upregulation of IQGAP1 affected the activity of Rap1 and the downstream phosphorylation of Src. In conclusion, these findings reveal an essential mechanism by which increased IQGAP1 in LPS-treated RPMVECs promotes barrier dysfunction and ICAM-1 upregulation, at least in part by regulating Rap1/Src signalling, indicating that IQGAP1 may be a potential therapeutic target to prevent endothelial hyperpermeability and inflammation in ALI.
Collapse
Affiliation(s)
- Na Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, People's Republic of China
| | - Gang Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, People's Republic of China
| | - Gengyun Sun
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, People's Republic of China.
| |
Collapse
|
26
|
Clementi N, Ghosh S, De Santis M, Castelli M, Criscuolo E, Zanoni I, Clementi M, Mancini N. Viral Respiratory Pathogens and Lung Injury. Clin Microbiol Rev 2021; 34:e00103-20. [PMID: 33789928 PMCID: PMC8142519 DOI: 10.1128/cmr.00103-20] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Several viruses target the human respiratory tract, causing different clinical manifestations spanning from mild upper airway involvement to life-threatening acute respiratory distress syndrome (ARDS). As dramatically evident in the ongoing SARS-CoV-2 pandemic, the clinical picture is not always easily predictable due to the combined effect of direct viral and indirect patient-specific immune-mediated damage. In this review, we discuss the main RNA (orthomyxoviruses, paramyxoviruses, and coronaviruses) and DNA (adenoviruses, herpesviruses, and bocaviruses) viruses with respiratory tropism and their mechanisms of direct and indirect cell damage. We analyze the thin line existing between a protective immune response, capable of limiting viral replication, and an unbalanced, dysregulated immune activation often leading to the most severe complication. Our comprehension of the molecular mechanisms involved is increasing and this should pave the way for the development and clinical use of new tailored immune-based antiviral strategies.
Collapse
Affiliation(s)
- Nicola Clementi
- Laboratory of Microbiology and Virology, Vita-Salute San Raffaele University, Milan, Italy
- Laboratory of Microbiology and Virology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Sreya Ghosh
- Harvard Medical School, Boston Children's Hospital, Division of Immunology, Boston, Massachusetts, USA
| | - Maria De Santis
- Department of Rheumatology and Clinical Immunology, Humanitas Clinical and Research Center-IRCCS, Rozzano, Italy
| | - Matteo Castelli
- Laboratory of Microbiology and Virology, Vita-Salute San Raffaele University, Milan, Italy
| | - Elena Criscuolo
- Laboratory of Microbiology and Virology, Vita-Salute San Raffaele University, Milan, Italy
| | - Ivan Zanoni
- Harvard Medical School, Boston Children's Hospital, Division of Immunology, Boston, Massachusetts, USA
- Harvard Medical School, Boston Children's Hospital, Division of Gastroenterology, Boston, Massachusetts, USA
| | - Massimo Clementi
- Laboratory of Microbiology and Virology, Vita-Salute San Raffaele University, Milan, Italy
- Laboratory of Microbiology and Virology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Nicasio Mancini
- Laboratory of Microbiology and Virology, Vita-Salute San Raffaele University, Milan, Italy
- Laboratory of Microbiology and Virology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
27
|
Bayir E, Sendemir A. Role of Intermediate Filaments in Blood-Brain Barrier in Health and Disease. Cells 2021; 10:cells10061400. [PMID: 34198868 PMCID: PMC8226756 DOI: 10.3390/cells10061400] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 05/31/2021] [Accepted: 06/02/2021] [Indexed: 12/11/2022] Open
Abstract
The blood–brain barrier (BBB) is a highly selective cellular monolayer unique to the microvasculature of the central nervous system (CNS), and it mediates the communication of the CNS with the rest of the body by regulating the passage of molecules into the CNS microenvironment. Limitation of passage of substances through the BBB is mainly due to tight junctions (TJ) and adherens junctions (AJ) between brain microvascular endothelial cells. The importance of actin filaments and microtubules in establishing and maintaining TJs and AJs has been indicated; however, recent studies have shown that intermediate filaments are also important in the formation and function of cell–cell junctions. The most common intermediate filament protein in endothelial cells is vimentin. Vimentin plays a role in blood–brain barrier permeability in both cell–cell and cell–matrix interactions by affecting the actin and microtubule reorganization and by binding directly to VE-cadherin or integrin proteins. The BBB permeability increases due to the formation of stress fibers and the disruption of VE–cadherin interactions between two neighboring cells in various diseases, disrupting the fiber network of intermediate filament vimentin in different ways. Intermediate filaments may be long ignored key targets in regulation of BBB permeability in health and disease.
Collapse
Affiliation(s)
- Ece Bayir
- Ege University Central Research Test and Analysis Laboratory Application and Research Center (EGE-MATAL), Ege University, 35100 Izmir, Turkey;
| | - Aylin Sendemir
- Department of Bioengineering, Faculty of Engineering, Ege University, 35100 Izmir, Turkey
- Department of Biomedical Technologies, Graduate School of Natural and Applied Science, Ege University, 35100 Izmir, Turkey
- Correspondence: ; Tel.: +90-232-3114817
| |
Collapse
|
28
|
Kovacs-Kasa A, Kovacs L, Cherian-Shaw M, Patel V, Meadows ML, Fulton DJ, Su Y, Verin AD. Inhibition of Class IIa HDACs improves endothelial barrier function in endotoxin-induced acute lung injury. J Cell Physiol 2021; 236:2893-2905. [PMID: 32959895 PMCID: PMC9946131 DOI: 10.1002/jcp.30053] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 08/31/2020] [Accepted: 09/03/2020] [Indexed: 12/21/2022]
Abstract
Acute lung injury (ALI) is an acute inflammatory process arises from a wide range of lung insults. A major cause of ALI is dysfunction of the pulmonary vascular endothelial barrier but the mechanisms involved are incompletely understood. The therapeutic potential of histone deacetylase (HDAC) inhibitors for the treatment of cardiovascular and inflammatory diseases is increasingly apparent, but the mechanisms by which HDACs regulate pulmonary vascular barrier function remain to be resolved. We found that specific Class IIa HDACs inhibitor, TMP269, significantly attenuated the lipopolysaccharide (LPS)-induced human lung microvascular endothelial cells (HLMVEC) barrier compromise in vitro and improved vascular barrier integrity and lung function in murine model of ALI in vivo. TMP269 decreased LPS-induced myosin light chain phosphorylation suggesting the role for Class IIa HDACs in LPS-induced cytoskeleton reorganization. TMP269 did not affect microtubule structure and tubulin acetylation in contrast to the HDAC6-specific inhibitor, Tubastatin A suggesting that Class IIa HDACs and HDAC6 (Class IIb) regulate endothelial cytoskeleton and permeability via different mechanisms. Furthermore, LPS increased the expression of ArgBP2 which has recently been attributed to HDAC-mediated activation of Rho. Depletion of ArgBP2 abolished the ability of LPS to disrupt barrier function in HLMVEC and both TMP269 and Tubastatin A decreased the level of ArgBP2 expression after LPS stimulation suggesting that both Class IIa and IIb HDACs regulate endothelial permeability via ArgBP2-dependent mechanism. Collectively, our data strongly suggest that Class IIa HDACs are involved in LPS-induced ALI in vitro and in vivo via specific mechanism which involved contractile responses, but not microtubule reorganization.
Collapse
Affiliation(s)
- Anita Kovacs-Kasa
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Laszlo Kovacs
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Mary Cherian-Shaw
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Vijay Patel
- Department of Cardiothoracic Surgery, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Mary L. Meadows
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - David J. Fulton
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Yunchao Su
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
- Department of Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
- Research Service, Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, USA
| | - Alexander D. Verin
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
- Department of Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| |
Collapse
|
29
|
Karki P, Cha B, Zhang CO, Li Y, Ke Y, Promnares K, Kaibuchi K, Yoshimura A, Birukov KG, Birukova AA. Microtubule-dependent mechanism of anti-inflammatory effect of SOCS1 in endothelial dysfunction and lung injury. FASEB J 2021; 35:e21388. [PMID: 33724556 PMCID: PMC10069762 DOI: 10.1096/fj.202001477rr] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 12/21/2020] [Accepted: 01/07/2021] [Indexed: 12/15/2022]
Abstract
Suppressors of cytokine signaling (SOCS) provide negative regulation of inflammatory reaction. The role and precise cellular mechanisms of SOCS1 in control of endothelial dysfunction and barrier compromise associated with acute lung injury remain unexplored. Our results show that siRNA-mediated SOCS1 knockdown augmented lipopolysaccharide (LPS)-induced pulmonary endothelial cell (EC) permeability and enhanced inflammatory response. Consistent with in vitro data, EC-specific SOCS1 knockout mice developed more severe lung vascular leak and accumulation of inflammatory cells in bronchoalveolar lavage fluid. SOCS1 overexpression exhibited protective effects against LPS-induced endothelial permeability and inflammation, which were dependent on microtubule (MT) integrity. Biochemical and image analysis of unstimulated EC showed SOCS1 association with the MT, while challenge with LPS or MT depolymerizing agent colchicine impaired this association. SOCS1 directly interacted with N2 domains of MT-associated proteins CLIP-170 and CLASP2. Furthermore, N-terminal region of SOCS1 was indispensable for these interactions and SOCS1-ΔN mutant lacking N-terminal 59 amino acids failed to rescue LPS-induced endothelial dysfunction. Depletion of endogenous CLIP-170 or CLASP2 abolished SOCS1 interaction with Toll-like receptor-4 and Janus kinase-2 leading to impairment of SOCS1 inhibitory effects on LPS-induced inflammation. Altogether, these findings suggest that endothelial barrier protective and anti-inflammatory effects of SOCS1 are critically dependent on its targeting to the MT.
Collapse
Affiliation(s)
- Pratap Karki
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Boyoung Cha
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Chen-Ou Zhang
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Yue Li
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Yunbo Ke
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kamoltip Promnares
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kozo Kaibuchi
- Department of Cell Pharmacology, Nagoya University, Nagoya, Japan
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University, Tokyo, Japan
| | - Konstantin G Birukov
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Anna A Birukova
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
30
|
Dragoni S, Caridi B, Karatsai E, Burgoyne T, Sarker MH, Turowski P. AMP-activated protein kinase is a key regulator of acute neurovascular permeability. J Cell Sci 2021; 134:jcs253179. [PMID: 33712448 PMCID: PMC8077405 DOI: 10.1242/jcs.253179] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 03/02/2021] [Indexed: 12/11/2022] Open
Abstract
Many neuronal and retinal disorders are associated with pathological hyperpermeability of the microvasculature. We have used explants of rodent retinae to study acute neurovascular permeability, signal transduction and the role of AMP-activated protein kinase (AMPK). Following stimulation with either vascular endothelial growth factor (VEGF-A) or bradykinin (BK), AMPK was rapidly and strongly phosphorylated and acted as a key mediator of permeability downstream of Ca2+. Accordingly, AMPK agonists potently induced acute retinal vascular leakage. AMPK activation led to phosphorylation of endothelial nitric oxide synthase (eNOS, also known as NOS3), which in turn increased VE-cadherin (CDH5) phosphorylation on Y685. In parallel, AMPK also mediated phosphorylation of p38 MAP kinases (hereafter p38) and HSP27 (HSPB1), indicating that it regulated paracellular junctions and cellular contractility, both previously associated with endothelial permeability. Endothelial AMPK provided a missing link in neurovascular permeability, connecting Ca2+ transients to the activation of eNOS and p38, irrespective of the permeability-inducing factor used. Collectively, we find that, due to its compatibility with small molecule antagonists and agonists, as well as siRNA, the ex vivo retina model constitutes a reliable tool to identify and study regulators and mechanisms of acute neurovascular permeability.
Collapse
Affiliation(s)
- Silvia Dragoni
- Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Bruna Caridi
- Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Eleni Karatsai
- Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Thomas Burgoyne
- Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Mosharraf H. Sarker
- Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
- School of Science, Engineering & Design, Teesside University, Stephenson Street, Middlesbrough TS1 3BA, UK
| | - Patric Turowski
- Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| |
Collapse
|
31
|
3D Cell-Culture Models for the Assessment of Anticoagulant and Anti-Inflammatory Properties of Endothelial Cells. Methods Mol Biol 2021; 2110:83-97. [PMID: 32002903 DOI: 10.1007/978-1-0716-0255-3_6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2023]
Abstract
Endothelial cells (EC) play a crucial role in the pathophysiology of cardiovascular diseases, ischemia/reperfusion injury, and graft rejection in (xeno-)transplantation. In such nonphysiological conditions, EC are known to lose their quiescent phenotype and switch into an actively pro-inflammatory, procoagulant, and anti-fibrinolytic state. This case happens essentially because the endothelial glycocalyx-a layer of proteoglycans and glycoproteins covering the luminal surface of the endothelium-is shed. Heparan sulfate, one of the main components of the endothelial glycocalyx, contributes to its negative charge. In addition, many plasma proteins such as antithrombin III, superoxide dismutase, C1 inhibitor, and growth factors and cytokines bind to heparan sulfate and by this scenario contribute to the establishment of an anticoagulant and anti-inflammatory endothelial surface. Shedding of the glycocalyx results in a loss of plasma proteins from the endothelial surface, and this phenomenon causes the switch in phenotype. Particularly in xenotransplantation, both hyperacute and acute vascular rejection are characterized by coagulation dysregulation, a situation in which EC are the main players.Since many years, EC have been used in vitro in 2D flatbed cell culture models, with or without the application of shear stress. Such models have also been used to assess the effect of human transgenes on complement- and coagulation-mediated damage of porcine EC in the context of xenotransplantation. The methods described in this chapter include the analysis of endothelial cell-blood interactions without the necessity of using anticoagulants as the increased EC surface-to-volume ratio allows for natural anticoagulation of blood. Furthermore, this chapter contains the description of a novel microfluidic in vitro model carrying important features of small blood vessels, such as a 3D round-section geometry, shear stress, and pulsatile flow-all this in a closed circuit, recirculating system aiming at reproducing closely the in vivo situation in small vessels.
Collapse
|
32
|
Chang R, Mamun A, Dominic A, Le NT. SARS-CoV-2 Mediated Endothelial Dysfunction: The Potential Role of Chronic Oxidative Stress. Front Physiol 2021; 11:605908. [PMID: 33519510 PMCID: PMC7844210 DOI: 10.3389/fphys.2020.605908] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 12/09/2020] [Indexed: 01/08/2023] Open
Abstract
Endothelial cells have emerged as key players in SARS-CoV-2 infection and COVID-19 inflammatory pathologies. Dysfunctional endothelial cells can promote chronic inflammation and disease processes like thrombosis, atherosclerosis, and lung injury. In endothelial cells, mitochondria regulate these inflammatory pathways via redox signaling, which is primarily achieved through mitochondrial reactive oxygen species (mtROS). Excess mtROS causes oxidative stress that can initiate and exacerbate senescence, a state that promotes inflammation and chronic endothelial dysfunction. Oxidative stress can also activate feedback loops that perpetuate mitochondrial dysfunction, mtROS overproduction, and inflammation. In this review, we provide an overview of phenotypes mediated by mtROS in endothelial cells - such as mitochondrial dysfunction, inflammation, and senescence - as well as how these chronic states may be initiated by SARS-CoV-2 infection of endothelial cells. We also propose that SARS-CoV-2 activates mtROS-mediated feedback loops that cause long-term changes in host redox status and endothelial function, promoting cardiovascular disease and lung injury after recovery from COVID-19. Finally, we discuss the implications of these proposed pathways on long-term vascular health and potential treatments to address these chronic conditions.
Collapse
Affiliation(s)
- Ryan Chang
- College of Arts & Sciences, Washington University in St. Louis, St. Louis, MO, United States
| | - Abrar Mamun
- Wiess School of Natural Sciences, Rice University, Houston, TX, United States
| | - Abishai Dominic
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University, College Station, TX, United States
- Department of Cardiovascular Sciences, Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, United States
| | - Nhat-Tu Le
- Department of Cardiovascular Sciences, Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, United States
| |
Collapse
|
33
|
Bossardi Ramos R, Adam AP. Molecular Mechanisms of Vascular Damage During Lung Injury. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1304:95-107. [PMID: 34019265 DOI: 10.1007/978-3-030-68748-9_6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A variety of pulmonary and systemic insults promote an inflammatory response causing increased vascular permeability, leading to the development of acute lung injury (ALI), a condition necessitating hospitalization and intensive care, or the more severe acute respiratory distress syndrome (ARDS), a disease with a high mortality rate. Further, COVID-19 pandemic-associated ARDS is now a major cause of mortality worldwide. The pathogenesis of ALI is explained by injury to both the vascular endothelium and the alveolar epithelium. The disruption of the lung endothelial and epithelial barriers occurs in response to both systemic and local production of pro-inflammatory cytokines. Studies that evaluate the association of genetic polymorphisms with disease risk did not yield many potential therapeutic targets to treat and revert lung injury. This failure is probably due in part to the phenotypic complexity of ALI/ARDS, and genetic predisposition may be obscured by the multiple environmental and behavioral risk factors. In the last decade, new research has uncovered novel epigenetic mechanisms that control ALI/ARDS pathogenesis, including histone modifications and DNA methylation. Enzyme inhibitors such as DNMTi and HDACi may offer new alternative strategies to prevent or reverse the vascular damage that occurs during lung injury. This review will focus on the latest findings on the molecular mechanisms of vascular damage in ALI/ARDS, the genetic factors that might contribute to the susceptibility for developing this disease, and the epigenetic changes observed in humans, as well as in experimental models of ALI/ADRS.
Collapse
Affiliation(s)
- Ramon Bossardi Ramos
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA.
| | - Alejandro Pablo Adam
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA. .,Department of Ophthalmology, Albany Medical College, Albany, NY, USA.
| |
Collapse
|
34
|
Karki P, Birukov KG. Oxidized Phospholipids in Control of Endothelial Barrier Function: Mechanisms and Implication in Lung Injury. Front Endocrinol (Lausanne) 2021; 12:794437. [PMID: 34887839 PMCID: PMC8649713 DOI: 10.3389/fendo.2021.794437] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 11/05/2021] [Indexed: 01/25/2023] Open
Abstract
Earlier studies investigating the pathogenesis of chronic vascular inflammation associated with atherosclerosis described pro-inflammatory and vascular barrier disruptive effects of lipid oxidation products accumulated in the sites of vascular lesion and atherosclerotic plaque. However, accumulating evidence including studies from our group suggests potent barrier protective and anti-inflammatory properties of certain oxidized phospholipids (OxPLs) in the lung vascular endothelium. Among these OxPLs, oxidized 1-palmitoyl-2-arachdonyl-sn-glycero-3-phosphocholine (OxPAPC) causes sustained enhancement of lung endothelial cell (EC) basal barrier properties and protects against vascular permeability induced by a wide variety of agonists ranging from bacterial pathogens and their cell wall components, endotoxins, thrombin, mechanical insults, and inflammatory cytokines. On the other hand, truncated OxPLs cause acute endothelial barrier disruption and potentiate inflammation. It appears that multiple signaling mechanisms triggering cytoskeletal remodeling are involved in OxPLs-mediated regulation of EC barrier. The promising vascular barrier protective and anti-inflammatory properties exhibited by OxPAPC and its particular components that have been established in the cellular and animal models of sepsis and acute lung injury has prompted consideration of OxPAPC as a prototype therapeutic molecule. In this review, we will summarize signaling and cytoskeletal mechanisms involved in OxPLs-mediated damage, rescue, and restoration of endothelial barrier in various pathophysiological settings and discuss a future potential of OxPAPC in treating lung disorders associated with endothelial barrier dysfunction.
Collapse
Affiliation(s)
- Pratap Karki
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Konstantin G. Birukov
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, United States
- *Correspondence: Konstantin G. Birukov,
| |
Collapse
|
35
|
Dolmatova EV, Wang K, Mandavilli R, Griendling KK. The effects of sepsis on endothelium and clinical implications. Cardiovasc Res 2021; 117:60-73. [PMID: 32215570 PMCID: PMC7810126 DOI: 10.1093/cvr/cvaa070] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 02/03/2020] [Accepted: 03/20/2020] [Indexed: 12/15/2022] Open
Abstract
ABSTRACT Sepsis accounts for nearly 700 000 deaths in Europe annually and is caused by an overwhelming host response to infection resulting in organ failure. The endothelium is an active contributor to sepsis and as such represents a major target for therapy. During sepsis, endothelial cells amplify the immune response and activate the coagulation system. They are both a target and source of inflammation and serve as a link between local and systemic immune responses. In response to cytokines produced by immune cells, the endothelium expresses adhesion molecules and produces vasoactive compounds, inflammatory cytokines, and chemoattractants, thus switching from an anticoagulant to procoagulant state. These responses contribute to local control of infection, but systemic activation can lead to microvascular thrombosis, capillary permeability, hypotension, tissue hypoxia, and ultimately tissue damage. This review focuses on the role of the endothelium in leucocyte adhesion and transmigration as well as production of reactive oxygen and nitrogen species, microRNAs and cytokines, formation of signalling microparticles, and disseminated intravascular coagulation. We also discuss alterations in endothelial permeability and apoptosis. Finally, we review the diagnostic potential of endothelial markers and endothelial pathways as therapeutic targets for this devastating disease.
Collapse
Affiliation(s)
- Elena V Dolmatova
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | - Keke Wang
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | - Rohan Mandavilli
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | - Kathy K Griendling
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, Atlanta, GA 30322, USA
| |
Collapse
|
36
|
Eisenhut M, Shin JI. Pathways in the Pathophysiology of Coronavirus 19 Lung Disease Accessible to Prevention and Treatment. Front Physiol 2020; 11:872. [PMID: 32922301 PMCID: PMC7457053 DOI: 10.3389/fphys.2020.00872] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 06/29/2020] [Indexed: 12/12/2022] Open
Abstract
Background: In COVID 19 related lung disease, which is a leading cause of death from this disease, cytokines like tumor necrosis factor-alpha (TNF alpha) may be pivotal in the pathogenesis. TNF alpha reduces fluid absorption due to impairment of sodium and chloride transport required for building an osmotic gradient across epithelial cells, which in the airways maintains airway surface liquid helping to keep airways open and enabling bacterial clearance and aids water absorption from the alveolar spaces. TNF alpha can, through Rho-kinase, disintegrate the endothelial and epithelial cytoskeleton, and thus break up intercellular tight junctional proteins, breaching the intercellular barrier, which prevents flooding of the interstitial and alveolar spaces with fluid. Hypotheses: (1) Preservation and restoration of airway and alveolar epithelial sodium and chloride transport and the cytoskeleton dependent integrity of the cell barriers within the lung can prevent and treat COVID 19 lung disease. (2) TNF alpha is the key mediator of pulmonary edema in COVID 19 lung disease. Confirmation of hypothesis and implications: The role of a reduction in the function of epithelial sodium and chloride transport could with regards to chloride transport be tested by analysis of chloride levels in exhaled breath condensate and levels correlated with TNF alpha concentrations. Reduced levels would indicate a reduction of the function of the cystic fibrosis transmembrane conductance regulator (CFTR) chloride channel and a correlation with TNF alpha levels indicative of its involvement. Anti-TNF alpha treatment with antibodies is already available and needs to be tested in randomized controlled trials of COVID 19 lung disease. TNF alpha levels could also be reduced by statins, aspirin, and curcumin. Chloride transport could be facilitated by CFTR activators, including curcumin and phosphodiesterase-5 inhibitors. Sodium and chloride transport could be further regulated to prevent accumulation of alveolar fluid by use of Na(+)/K(+)/2Cl(-) cotransporter type 1 inhibitors, which have been associated with improved outcome in adults ventilated for acute respiratory distress syndrome (ARDS) in randomized controlled trials. Primary prevention of coronavirus infection and TNF alpha release in response to it could be improved by induction of antimicrobial peptides LL-37 and human beta defensin-2 and reduction of TNF alpha production by vitamin D prophylaxis for the population as a whole.
Collapse
Affiliation(s)
- Michael Eisenhut
- Children's & Adolescent Services, Luton & Dunstable University Hospital NHS Foundation Trust, Luton, United Kingdom
| | - Jae Il Shin
- Department of Pediatrics, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
37
|
Wang Y, Wang M, Yu P, Zuo L, Zhou Q, Zhou X, Zhu H. MicroRNA-126 Modulates Palmitate-Induced Migration in HUVECs by Downregulating Myosin Light Chain Kinase via the ERK/MAPK Pathway. Front Bioeng Biotechnol 2020; 8:913. [PMID: 32850751 PMCID: PMC7411007 DOI: 10.3389/fbioe.2020.00913] [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: 05/29/2020] [Accepted: 07/15/2020] [Indexed: 12/23/2022] Open
Abstract
MicroRNA-126 (miR-126) is an endothelial-specific microRNA that has shown beneficial effects on endothelial dysfunction. However, the underlying molecular mechanism is unclear. The present study evaluated the effects of miR-126 on the cell migration and underlying mechanism in HUVECs treated with palmitate. The present results demonstrated that overexpression of miR-126 was found to decrease cell migration in palmitate-treated HUVECs, with decreased MLCK expression and subsequent decreased phosphorylated MLC level. miR-126 also decreased the phosphorylation of MYPT1 in palmitate-treated HUVECs. In addition, it was demonstrated that miR-126 decreases expression of the NADPH oxidase subunits, p67 and Rac family small GTPase 1 with a subsequent decrease in cell apoptosis. Moreover, the phosphorylation of ERK was reduced by miR-126 in palmitate-induced HUVECs. Taken together, the present study showed that the effect of miR-126 on cell migration and cell apoptosis is mediated through downregulation of MLCK via the ERK/MAPK pathway.
Collapse
Affiliation(s)
- Yi Wang
- Department of Biological Engineering, School of Life Sciences, Anhui Medical University, Hefei, China.,Laboratory of Molecular Biology and Department of Biochemistry, Anhui Medical University, Hefei, China
| | - Mei Wang
- General Department of Hyperbaric Oxygen, Hefei Hospital Affiliated to Anhui Medical University, Hefei, China
| | - Pei Yu
- Laboratory of Molecular Biology and Department of Biochemistry, Anhui Medical University, Hefei, China
| | - Li Zuo
- Laboratory of Molecular Biology and Department of Biochemistry, Anhui Medical University, Hefei, China
| | - Qing Zhou
- Laboratory of Molecular Biology and Department of Biochemistry, Anhui Medical University, Hefei, China
| | - Xiaomei Zhou
- General Department of Hyperbaric Oxygen, Hefei Hospital Affiliated to Anhui Medical University, Hefei, China
| | - Huaqing Zhu
- Laboratory of Molecular Biology and Department of Biochemistry, Anhui Medical University, Hefei, China
| |
Collapse
|
38
|
Okamoto T, Kawamoto E, Usuda H, Tanaka T, Nikai T, Asanuma K, Suzuki K, Shimaoka M, Wada K. Recombinant Human Soluble Thrombomodulin Suppresses Monocyte Adhesion by Reducing Lipopolysaccharide-Induced Endothelial Cellular Stiffening. Cells 2020; 9:cells9081811. [PMID: 32751580 PMCID: PMC7463703 DOI: 10.3390/cells9081811] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/21/2020] [Accepted: 07/28/2020] [Indexed: 11/24/2022] Open
Abstract
Endothelial cellular stiffening has been observed not only in inflamed cultured endothelial cells but also in the endothelium of atherosclerotic regions, which is an underlying cause of monocyte adhesion and accumulation. Although recombinant soluble thrombomodulin (rsTM) has been reported to suppress the inflammatory response of endothelial cells, its role in regulating endothelial cellular stiffness remains unclear. The purpose of this study was to investigate the impact of anticoagulant rsTM on lipopolysaccharide (LPS)-induced endothelial cellular stiffening. We show that LPS increases endothelial cellular stiffness by using atomic force microscopy and that rsTM reduces LPS-induced cellular stiffening not only through the attenuation of actin fiber and focal adhesion formation but also via the improvement of gap junction functionality. Moreover, post-administration of rsTM, after LPS stimulation, attenuated LPS-induced cellular stiffening. We also found that endothelial cells regulate leukocyte adhesion in a substrate- and cellular stiffness-dependent manner. Our result show that LPS-induced cellular stiffening enhances monocytic THP-1 cell line adhesion, whereas rsTM suppresses THP-1 cell adhesion to inflamed endothelial cells by reducing cellular stiffness. Endothelial cells increase cellular stiffness in reaction to inflammation, thereby promoting monocyte adhesion. Treatment of rsTM reduced LPS-induced cellular stiffening and suppressed monocyte adhesion in a cellular stiffness-dependent manner.
Collapse
Affiliation(s)
- Takayuki Okamoto
- Department of Pharmacology, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo-city, Shimane 693-8501, Japan; (H.U.); (T.T.); (K.W.)
- Correspondence: ; Tel.: +81-853-20-2132
| | - Eiji Kawamoto
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-city, Mie 514-8507, Japan; (E.K.); (M.S.)
- Department of Emergency and Disaster Medicine, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-city, Mie 514-8507, Japan
| | - Haruki Usuda
- Department of Pharmacology, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo-city, Shimane 693-8501, Japan; (H.U.); (T.T.); (K.W.)
| | - Tetsuya Tanaka
- Department of Pharmacology, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo-city, Shimane 693-8501, Japan; (H.U.); (T.T.); (K.W.)
- Department of Human Nutrition, Faculty of Contemporary Life Science, Chugoku Gakuen University, 83 Niwase, Kita-ku, Okayama-city, Okayama 701-0197, Japan
| | - Tetsuro Nikai
- Department of Anesthesiology, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo-city, Shimane 693-8501, Japan;
| | - Kunihiro Asanuma
- Department of Orthopaedic Surgery, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-city, Mie 514-8507, Japan;
| | - Koji Suzuki
- Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, 3500-3, Minamitamagaki-cho, Suzuka-city, Mie 513-8679, Japan;
| | - Motomu Shimaoka
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-city, Mie 514-8507, Japan; (E.K.); (M.S.)
| | - Koichiro Wada
- Department of Pharmacology, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo-city, Shimane 693-8501, Japan; (H.U.); (T.T.); (K.W.)
| |
Collapse
|
39
|
Structural Integrity of the Alveolar-Capillary Barrier in Cynomolgus Monkeys Challenged with Fully Virulent and Toxin-Deficient Strains of Bacillus anthracis. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:2095-2110. [PMID: 32598882 DOI: 10.1016/j.ajpath.2020.06.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/27/2020] [Accepted: 06/09/2020] [Indexed: 11/21/2022]
Abstract
Inhalational anthrax, a disease caused by inhaling Bacillus anthracis spores, leads to respiratory distress, vascular leakage, high-level bacteremia, and often death within days. Anthrax lethal toxin and edema toxin, which are composed of protective antigen (PA) plus either lethal factor (LF) or edema factor (EF), respectively, play an important yet incompletely defined role in the pulmonary pathophysiology. To better understand their contribution, we examined the structural integrity of the alveolar-capillary barrier in archival formalin-fixed lungs of cynomolgus monkeys challenged with the fully virulent B. anthracis Ames wild-type strain or the isogenic toxin-deficient mutants ΔEF, ΔLF, and ΔPA. Pulmonary spore challenge with the wild-type strain caused high mortality, intra-alveolar hemorrhages, extensive alveolar septal sequestration of bacteria and neutrophils, diffuse destabilization of epithelial and endothelial junctions, increased markers of coagulation and complement activation (including tissue factor and C5a), and multifocal intra-alveolar fibrin deposition. ΔEF challenge was lethal and showed similar alveolar-capillary alterations; however, intra-alveolar hemorrhages, bacterial deposition, and markers of coagulation or complement were absent or markedly lower. In contrast, ΔLF or ΔPA challenges were nonlethal and showed no signs of alveolar bacterial deposition or alveolar-capillary changes. These findings provide evidence that lethal toxin plays a determinative role in bacterial dissemination and alveolar-capillary barrier dysfunction, and edema toxin may significantly exacerbate pulmonary pathologies in a systemic infection.
Collapse
|
40
|
Verin AD, Batori R, Kovacs-Kasa A, Cherian-Shaw M, Kumar S, Czikora I, Karoor V, Strassheim D, Stenmark KR, Gerasimovskaya EV. Extracellular adenosine enhances pulmonary artery vasa vasorum endothelial cell barrier function via Gi/ELMO1/Rac1/PKA-dependent signaling mechanisms. Am J Physiol Cell Physiol 2020; 319:C183-C193. [PMID: 32432925 DOI: 10.1152/ajpcell.00505.2019] [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] [Indexed: 12/20/2022]
Abstract
The vasa vasorum (VV), the microvascular network around large vessels, has been recognized as an important contributor to the pathological vascular remodeling in cardiovascular diseases. In bovine and rat models of hypoxic pulmonary hypertension (PH), we have previously shown that chronic hypoxia profoundly increased pulmonary artery (PA) VV permeability, associated with infiltration of inflammatory and progenitor cells in the arterial wall, perivascular inflammation, and structural vascular remodeling. Extracellular adenosine was shown to exhibit a barrier-protective effect on VV endothelial cells (VVEC) via cAMP-independent mechanisms, which involved adenosine A1 receptor-mediated activation of Gi-phosphoinositide 3-kinase-Akt pathway and actin cytoskeleton remodeling. Using VVEC isolated from the adventitia of calf PA, in this study we investigated in more detail the mechanisms linking Gi activation to downstream barrier protection pathways. Using a small-interference RNA (siRNA) technique and transendothelial electrical resistance assay, we found that the adaptor protein, engulfment and cell motility 1 (ELMO1), the tyrosine phosphatase Src homology region 2 domain-containing phosphatase-2, and atypical Gi- and Rac1-mediated protein kinase A activation are implicated in VVEC barrier enhancement. In contrast, the actin-interacting GTP-binding protein, girdin, and the p21-activated kinase 1 downstream target, LIM kinase, are not involved in this response. In addition, adenosine-dependent cytoskeletal rearrangement involves activation of cofilin and inactivation of ezrin-radixin-moesin regulatory cytoskeletal proteins, consistent with a barrier-protective mechanism. Collectively, our data indicate that targeting adenosine receptors and downstream barrier-protective pathways in VVEC may have a potential translational significance in developing pharmacological approach for the VV barrier protection in PH.
Collapse
Affiliation(s)
| | - Robert Batori
- Augusta University Vascular Biology Center, Augusta, Georgia
| | | | | | - Sanjiv Kumar
- Augusta University Vascular Biology Center, Augusta, Georgia
| | - Istvan Czikora
- Augusta University Vascular Biology Center, Augusta, Georgia
| | - Vijaya Karoor
- Department of Medicine, University of Colorado Denver, Aurora, Colorado
| | - Derek Strassheim
- Department of Medicine, University of Colorado Denver, Aurora, Colorado
| | - Kurt R Stenmark
- Department of Pediatrics, University of Colorado Denver, Aurora, Colorado
| | | |
Collapse
|
41
|
van den Bos R, Cromwijk S, Tschigg K, Althuizen J, Zethof J, Whelan R, Flik G, Schaaf M. Early Life Glucocorticoid Exposure Modulates Immune Function in Zebrafish ( Danio rerio) Larvae. Front Immunol 2020; 11:727. [PMID: 32411141 PMCID: PMC7201046 DOI: 10.3389/fimmu.2020.00727] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 03/31/2020] [Indexed: 11/17/2022] Open
Abstract
In this study we have assessed the effects of increased cortisol levels during early embryonic development on immune function in zebrafish (Danio rerio) larvae. Fertilized eggs were exposed to either a cortisol-containing, a dexamethasone-containing (to stimulate the glucocorticoid receptor selectively) or a control medium for 6 h post-fertilization (0–6 hpf). First, we measured baseline expression of a number of immune-related genes (socs3a, mpeg1.1, mpeg1.2, and irg1l) 5 days post-fertilization (dpf) in larvae of the AB and TL strain to assess the effectiveness of our exposure procedure and potential strain differences. Cortisol and dexamethasone strongly up-regulated baseline expression of these genes independent of strain. The next series of experiments were therefore carried out in larvae of the AB strain only. We measured neutrophil/macrophage recruitment following tail fin amputation (performed at 3 dpf) and phenotypical changes as well as survival following LPS-induced sepsis (150 μg/ml; 4–5 dpf). Dexamethasone, but not cortisol, exposure at 0–6 hpf enhanced neutrophil recruitment 4 h post tail fin amputation. Cortisol and dexamethasone exposure at 0–6 hpf led to a milder phenotype (e.g., less tail fin damage) and enhanced survival following LPS challenge compared to control exposure. Gene-expression analysis showed accompanying differences in transcript abundance of tlr4bb, cxcr4a, myd88, il1β, and il10. These data show that early-life exposure to cortisol, which may be considered to be a model or proxy of maternal stress, induces an adaptive response to immune challenges, which seems mediated via the glucocorticoid receptor.
Collapse
Affiliation(s)
- Ruud van den Bos
- Department of Animal Ecology and Physiology, Institute of Water and Wetland Research, Faculty of Science, Radboud University, Nijmegen, Netherlands
| | - Suzanne Cromwijk
- Department of Animal Ecology and Physiology, Institute of Water and Wetland Research, Faculty of Science, Radboud University, Nijmegen, Netherlands
| | - Katharina Tschigg
- Department of Animal Ecology and Physiology, Institute of Water and Wetland Research, Faculty of Science, Radboud University, Nijmegen, Netherlands
| | - Joep Althuizen
- Department of Animal Ecology and Physiology, Institute of Water and Wetland Research, Faculty of Science, Radboud University, Nijmegen, Netherlands
| | - Jan Zethof
- Department of Animal Ecology and Physiology, Institute of Water and Wetland Research, Faculty of Science, Radboud University, Nijmegen, Netherlands
| | - Robert Whelan
- Animal Sciences and Health Cluster, Institute of Biology, Leiden University, Leiden, Netherlands
| | - Gert Flik
- Department of Animal Ecology and Physiology, Institute of Water and Wetland Research, Faculty of Science, Radboud University, Nijmegen, Netherlands
| | - Marcel Schaaf
- Animal Sciences and Health Cluster, Institute of Biology, Leiden University, Leiden, Netherlands
| |
Collapse
|
42
|
Karki P, Birukov KG. Oxidized Phospholipids in Healthy and Diseased Lung Endothelium. Cells 2020; 9:cells9040981. [PMID: 32326516 PMCID: PMC7226969 DOI: 10.3390/cells9040981] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/08/2020] [Accepted: 04/10/2020] [Indexed: 12/11/2022] Open
Abstract
Circulating and cell membrane phospholipids undergo oxidation caused by enzymatic and non-enzymatic mechanisms. As a result, a diverse group of bioactive oxidized phospholipids generated in these conditions have both beneficial and harmful effects on the human body. Increased production of oxidized phospholipid products with deleterious effects is linked to the pathogenesis of various cardiopulmonary disorders such as atherosclerosis, thrombosis, acute lung injury (ALI), and inflammation. It has been determined that the contrasting biological effects of lipid oxidation products are governed by their structural variations. For example, full-length products of 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphorylcholine oxidation (OxPAPC) have prominent endothelial barrier protective and anti-inflammatory activities while most of the truncated oxidized phospholipids induce vascular leak and exacerbate inflammation. The extensive studies from our group and other groups have demonstrated a strong potential of OxPAPC in mitigating a wide range of agonist-induced lung injuries and inflammation in pulmonary endothelial cell culture and rodent models of ALI. Concurrently, elevated levels of truncated oxidized phospholipids are present in aged mice lungs that potentiate the inflammatory agents-induced lung injury. On the other hand, increased levels of full length OxPAPC products accelerate ALI recovery by facilitating production of anti-inflammatory lipid mediator, lipoxin A4, and other molecules with anti-inflammatory properties. These findings suggest that OxPAPC-assisted lipid program switch may be a promising therapeutic strategy for treatment of acute inflammatory syndromes. In this review, we will summarize the vascular-protective and deleterious aspects of oxidized phospholipids and discuss their therapeutic potential including engineering of stable analogs of oxidized phospholipids with improved anti-inflammatory and barrier-protective properties.
Collapse
Affiliation(s)
- Pratap Karki
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | - Konstantin G. Birukov
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Correspondence: ; Tel.: +1-(410)-706-2578; Fax: +1-(410)-706-6952
| |
Collapse
|
43
|
Bowyer JF, Sarkar S, Burks SM, Hess JN, Tolani S, O'Callaghan JP, Hanig JP. Microglial activation and responses to vasculature that result from an acute LPS exposure. Neurotoxicology 2020; 77:181-192. [PMID: 32014511 DOI: 10.1016/j.neuro.2020.01.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 01/29/2020] [Accepted: 01/31/2020] [Indexed: 12/22/2022]
Abstract
Bacterial cell wall endotoxins, i.e. lipopolysaccharides (LPS), are some of the original compounds shown to evoke the classic signs of systemic inflammation/innate immune response and neuroinflammation. The term neuroinflammation often is used to infer the elaboration of proinflammatory mediators by microglia elicited by neuronal targeted activity. However, it also is possible that the microglia are responding to vasculature through several signaling mechanisms. Microglial activation relative to the vasculature in the hippocampus and parietal cortex was determined after an acute exposure of a single subcutaneous injection of 2 mg/kg LPS. Antibodies to allograft inflammatory factor (Aif1, a.k.a. Iba1) were used to track and quantify morphological changes in microglia. Immunostaining of platelet/endothelial cell adhesion molecule 1 (Pecam1, a.k.a. Cd31) was used to visualize vasculature in the forebrain and glial acidic fibrillary protein (GFAP) to visualize astrocytes. Neuroinflammation and other aspects of neurotoxicity were evaluated histologically at 3 h, 6 h, 12 h, 24 h, 3 d and 14 d following LPS exposure. LPS did not cause neurodegeneration as determined by Fluoro Jade C labeling. Also, there were no signs of mouse IgG leakage from brain vasculature due to LPS. Some changes in microglia size occurred at 6 h, but by 12 h microglial activation had begun with the combined soma and proximal processes size increasing significantly (1.5-fold). At 24 h, almost all the microglia soma and proximal processes in the hippocampus, parietal cortex, and thalamus were closely associated with the vasculature and had increased almost 2.0-fold in size. In many areas where microglia were juxtaposed to vasculature, astrocytic endfeet appeared to be displaced. The microglial activation had subsided slightly by 3 d with microglial size 1.6-fold that of control. We hypothesize that acute LPS activation can result in vascular mediated microglial responses through several mechanisms: 1) binding to Cd14 and Tlr4 receptors on microglia processes residing on vasculature; 2) damaging vasculature and causing the release of cytokines; and 3) possibly astrocytic endfeet damage resulting in cytokine release. These acute responses may serve as an adaptive mechanism to exposure to circulating LPS where the microglia surround the vasculature. This could further prevent the pathogen(s) circulating in blood from entering the brain. However, diverting microglial interactions away from synaptic remodeling and other types of microglial interactions with neurons may have adverse effects on neuronal function.
Collapse
Affiliation(s)
- John F Bowyer
- Division of Neurotoxicology, National Center for Toxicology/ FDA, Jefferson, AR 72079, USA
| | - Sumit Sarkar
- Division of Neurotoxicology, National Center for Toxicology/ FDA, Jefferson, AR 72079, USA.
| | - Susan M Burks
- Division of Neurotoxicology, National Center for Toxicology/ FDA, Jefferson, AR 72079, USA
| | - Jade N Hess
- Division of Neurotoxicology, National Center for Toxicology/ FDA, Jefferson, AR 72079, USA
| | - Serena Tolani
- Division of Neurotoxicology, National Center for Toxicology/ FDA, Jefferson, AR 72079, USA
| | - James P O'Callaghan
- Health Effects Laboratory Division, Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health Morgantown, WV 26505, USA
| | - Joseph P Hanig
- Center for Drug Evaluation and Research/ FDA Silver Spring, MD 20993, USA
| |
Collapse
|
44
|
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: 35] [Impact Index Per Article: 5.8] [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
|
45
|
Herrero-Fernandez B, Gomez-Bris R, Somovilla-Crespo B, Gonzalez-Granado JM. Immunobiology of Atherosclerosis: A Complex Net of Interactions. Int J Mol Sci 2019; 20:E5293. [PMID: 31653058 PMCID: PMC6862594 DOI: 10.3390/ijms20215293] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease is the leading cause of mortality worldwide, and atherosclerosis the principal factor underlying cardiovascular events. Atherosclerosis is a chronic inflammatory disease characterized by endothelial dysfunction, intimal lipid deposition, smooth muscle cell proliferation, cell apoptosis and necrosis, and local and systemic inflammation, involving key contributions to from innate and adaptive immunity. The balance between proatherogenic inflammatory and atheroprotective anti-inflammatory responses is modulated by a complex network of interactions among vascular components and immune cells, including monocytes, macrophages, dendritic cells, and T, B, and foam cells; these interactions modulate the further progression and stability of the atherosclerotic lesion. In this review, we take a global perspective on existing knowledge about the pathogenesis of immune responses in the atherosclerotic microenvironment and the interplay between the major innate and adaptive immune factors in atherosclerosis. Studies such as this are the basis for the development of new therapies against atherosclerosis.
Collapse
Affiliation(s)
- Beatriz Herrero-Fernandez
- LamImSys Lab. Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain.
- Departamento de Fisiología. Facultad de Medicina. Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain.
| | - Raquel Gomez-Bris
- LamImSys Lab. Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain.
| | | | - Jose Maria Gonzalez-Granado
- LamImSys Lab. Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain.
- Departamento de Fisiología. Facultad de Medicina. Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain.
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain.
- CIBER de Enfermedades Cardiovasculares, 28029 Madrid, Spain.
| |
Collapse
|
46
|
Engler AJ, Raredon MSB, Le AV, Yuan Y, Oczkowicz YA, Kan EL, Baevova P, Niklason LE. Non-invasive and real-time measurement of microvascular barrier in intact lungs. Biomaterials 2019; 217:119313. [PMID: 31280072 PMCID: PMC6863174 DOI: 10.1016/j.biomaterials.2019.119313] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 06/25/2019] [Accepted: 06/27/2019] [Indexed: 12/15/2022]
Abstract
Microvascular leak is a phenomenon witnessed in multiple disease states. In organ engineering, regaining a functional barrier is the most crucial step towards creating an implantable organ. All previous methods of measuring microvascular permeability were either invasive, lengthy, introduced exogenous macromolecules, or relied on extrapolations from cultured cells. We present here a system that enables real-time measurement of microvascular permeability in intact rat lungs. Our unique system design allows direct, non-invasive measurement of average alveolar and capillary pressures, tracks flow paths within the organ, and enables calculation of lumped internal resistances including microvascular barrier. We first describe the physiology of native and decellularized lungs and the inherent properties of the extracellular matrix as functions of perfusion rate. We next track changing internal resistances and flows in injured native rat lungs, resolving the onset of microvascular leak, quantifying changing vascular resistances, and identifying distinct phases of organ failure. Finally, we measure changes in permeability within engineered lungs seeded with microvascular endothelial cells, quantifying cellular effects on internal vascular and barrier resistances over time. This system marks considerable progress in bioreactor design for intact organs and may be used to monitor and garner physiological insights into native, decellularized, and engineered tissues.
Collapse
Affiliation(s)
- Alexander J Engler
- Department of Biomedical Engineering, Yale University School of Engineering and Applied Science, New Haven, CT, USA
| | - Micha Sam B Raredon
- Department of Biomedical Engineering, Yale University School of Engineering and Applied Science, New Haven, CT, USA
| | - Andrew V Le
- Department of Anesthesiology, Yale University School of Medicine, New Haven, CT, USA
| | - Yifan Yuan
- Department of Anesthesiology, Yale University School of Medicine, New Haven, CT, USA
| | - Yan A Oczkowicz
- Department of Biomedical Engineering, Yale University School of Engineering and Applied Science, New Haven, CT, USA
| | - Ellen L Kan
- Department of Biomedical Engineering, Yale University School of Engineering and Applied Science, New Haven, CT, USA
| | - Pavlina Baevova
- Department of Anesthesiology, Yale University School of Medicine, New Haven, CT, USA
| | - Laura E Niklason
- Department of Biomedical Engineering, Yale University School of Engineering and Applied Science, New Haven, CT, USA; Department of Anesthesiology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
47
|
Wu D, Birukov K. Endothelial Cell Mechano-Metabolomic Coupling to Disease States in the Lung Microvasculature. Front Bioeng Biotechnol 2019; 7:172. [PMID: 31380363 PMCID: PMC6658821 DOI: 10.3389/fbioe.2019.00172] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/03/2019] [Indexed: 12/15/2022] Open
Abstract
Lungs are the most vascular part of humans, accepting the totality of cardiac output in a volume much smaller than the body itself. Due to this cardiac output, the lung microvasculature is subject to mechanical forces including shear stress and cyclic stretch that vary with the cardiac and breathing cycle. Vessels are surrounded by extracellular matrix which dictates the stiffness which endothelial cells also sense and respond to. Shear stress, stiffness, and cyclic stretch are known to influence endothelial cell state. At high shear stress, endothelial cells exhibit cell quiescence marked by low inflammatory markers and high nitric oxide synthesis, whereas at low shear stress, endothelial cells are thought to "activate" into a pro-inflammatory state and have low nitric oxide. Shear stress' profound effect on vascular phenotype is most apparent in the arterial vasculature and in the pathophysiology of vascular inflammation. To conduct the flow of blood from the right heart, the lung microvasculature must be rigid yet compliant. It turns out that excessive substrate rigidity or stiffness is important in the development of pulmonary hypertension and chronic fibrosing lung diseases via excessive cell proliferation or the endothelial-mesenchymal transition. Recently, a new body of literature has evolved that couples mechanical sensing to endothelial phenotypic changes through metabolic signaling in clinically relevant contexts such as pulmonary hypertension, lung injury syndromes, as well as fibrosis, which is the focus of this review. Stretch, like flow, has profound effect on endothelial phenotype; metabolism studies due to stretch are in their infancy.
Collapse
Affiliation(s)
- David Wu
- Section of Pulmonary and Critical Care, Department of Medicine, University of Chicago, Chicago, IL, United States
| | - Konstantin Birukov
- Department of Anesthesia, University of Maryland, Baltimore, MD, United States
| |
Collapse
|
48
|
Wang X, Obeidat M, Li L, Pasarj P, Aburahess S, Holmes CFB, Ballermann BJ. TIMAP inhibits endothelial myosin light chain phosphatase by competing with MYPT1 for the catalytic protein phosphatase 1 subunit PP1cβ. J Biol Chem 2019; 294:13280-13291. [PMID: 31315927 PMCID: PMC6737228 DOI: 10.1074/jbc.ra118.006075] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 06/17/2019] [Indexed: 11/06/2022] Open
Abstract
Transforming growth factor-β membrane associated protein (TIMAP) is an endothelial cell (EC)-predominant PP1 regulatory subunit and a member of the myosin phosphatase target (MYPT) protein family. The MYPTs preferentially bind the catalytic protein phosphatase 1 subunit PP1cβ, forming myosin phosphatase holoenzymes. We investigated whether TIMAP/PP1cβ could also function as a myosin phosphatase. Endogenous PP1cβ, myosin light chain 2 (MLC2), and myosin IIA heavy chain coimmunoprecipitated from EC lysates with endogenous TIMAP, and endogenous MLC2 colocalized with TIMAP in EC projections. Purified recombinant GST-TIMAP interacted directly with purified recombinant His-MLC2. However, TIMAP overexpression in EC enhanced MLC2 phosphorylation, an effect not observed with a TIMAP mutant that does not bind PP1cβ. Conversely, MLC2 phosphorylation was reduced in lung lysates from TIMAP-deficient mice and upon silencing of endogenous TIMAP expression in ECs. Ectopically expressed TIMAP slowed the rate of MLC2 dephosphorylation, an effect requiring TIMAP-PP1cβ interaction. The association of MYPT1 with PP1cβ was profoundly reduced in the presence of excess TIMAP, leading to proteasomal MYPT1 degradation. In the absence of TIMAP, MYPT1-associated PP1cβ readily bound immobilized microcystin-LR, an active-site inhibitor of PP1c. By contrast, TIMAP-associated PP1cβ did not interact with microcystin-LR, indicating that the active site of PP1cβ is blocked when it is bound to TIMAP. Thus, TIMAP inhibits myosin phosphatase activity in ECs by competing with MYPT1 for PP1cβ and blocking the PP1cβ active site.
Collapse
Affiliation(s)
- Xin Wang
- Department of Medicine, University of Alberta, Edmonton, Alberta T6G 2G3, Canada
| | - Marya Obeidat
- Department of Medicine, University of Alberta, Edmonton, Alberta T6G 2G3, Canada
| | - Laiji Li
- Department of Medicine, University of Alberta, Edmonton, Alberta T6G 2G3, Canada
| | - Phuwadet Pasarj
- Department of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2G3, Canada
| | - Salah Aburahess
- Department of Medicine, University of Alberta, Edmonton, Alberta T6G 2G3, Canada
| | - Charles F B Holmes
- Department of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2G3, Canada
| | - Barbara J Ballermann
- Department of Medicine, University of Alberta, Edmonton, Alberta T6G 2G3, Canada.
| |
Collapse
|
49
|
Abstract
BACKGROUND Microvascular hyperpermeability resulting from endothelial barrier dysfunction (EBD) is associated with worse clinical outcomes in trauma-induced hemorrhagic shock. We have previously shown that treatment with Tubastatin A (TubA), a histone deacetylase 6 inhibitor, improves outcomes in animal models of shock. In this study, we investigate whether TubA treatment may prevent trauma-related EBD. METHODS Wistar-Kyoto rats subjected to 40% hemorrhage were treated with TubA or vehicle control. Acute lung injury (ALI) was assessed histologically from tissues harvested 6 hours posthemorrhage. In vitro, human umbilical vein endothelial cells (HUVECs) were cultured in EGM BulletKit medium. Medium was exchanged for glucose-free Dulbecco's Modified Eagle Medium (0.5% fetal bovine serum) with or without TubA, and cells were placed in an anoxic chamber (5% CO2, 95% N2, 20-48 hours). Expression of acetylated tubulin and hypoxia-inducible factor 1α was measured by Western blot. Soluble Intercellular Adhesion Molecule-1 concentration within the medium, a marker of endothelial integrity, was determined using enzyme-linked immunosorbent assay. Monolayers were assessed for permeability via transwell assays using fluorescein isothiocyanate-labeled albumin. RESULTS Rats treated with TubA had significantly reduced ALI relative to vehicle control. In vitro, TubA significantly attenuated anoxia-induced hyperpermeability, hypoxia-inducible factor 1α expression, and glycocalyx shedding. CONCLUSIONS Our findings demonstrate that TubA prevents hemorrhage-induced ALI in rats. Additionally, we have shown that TubA prevents anoxia-induced EBD in vitro. Taken together, these results suggest that TubA could attenuate microvascular hyperpermeability related to hemorrhagic shock.
Collapse
|
50
|
Smirnova SS, Pisareva MM, Smirnova TD, Sivak KV, Vorobiev KV. Long-Term Maintenance of the Functional Changes Induced by Influenza A Virus and/or LPS in Human Endothelial ECV-304 Cell Sublines. ACTA ACUST UNITED AC 2019; 13:283-291. [PMID: 32288938 PMCID: PMC7101551 DOI: 10.1134/s1990519x19040084] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 12/16/2018] [Accepted: 12/25/2018] [Indexed: 11/23/2022]
Abstract
Influenza A virus and secondary bacterial infection may have remote effects in the form of cardiovascular complications or fibrosis in different organs. However, the mechanisms governing the development of complications remain poorly studied. The present work reports the comparative assessment of the functional changes which take place in human ECV-304 endothelial cell sublines obtained previously by the long-term culturing of cells after exposure to varying infectious doses (IDs) of influenza A virus, and/or bacterial lipopolysaccharide (LPS). It has been demonstrated that, in the course of long-term culturing (six passages) after exposure to pathogenic agents (influenza virus and/or LPS), endothelial cells maintain changes in their migratory activity, permeability, and expression of mRNA for cytokines TNFα and TGFβ (along with the changes in their proliferation activity, which has been demonstrated earlier). The pattern of changes depended on the type of the agent (agents) to which the cells were exposed. The differences in migratory activity (which was at its maximum 4 h after wounding) between the cell sublines at the sixth passage correlated with the differences in their proliferation activity at the first passage (proliferation data were obtained previously). In particular, an increase in migration and proliferation was observed in the sublines exposed to low virus doses (ECV-1ID), as well as exposed to LPS (ECV-LPS), while the suppression of migration and proliferation was observed in the subline exposed to high virus doses (ECV-1000ID). In the ECV-1ID, ECV-LPS, and most notably in ECV-1ID + LPS sublines, we detected an increase in the expression of mRNA for cytokines TNFα and TGFβ, which, however, didn’t lead to the induction of apoptosis. We have also demonstrated an increase in cell permeability in the analyzed sublines, which was indicated by a decrease in the expression of the mRNAs for the genes encoding occludin and ZO-1, the tight junctions proteins . This paper also reports an evaluation of the effects of the antiviral preparations rimantadine and alpisarin on the functional state of cell sublines. As a result, it has been demonstrated that these drugs may be able to prevent the development of the pathological changes caused by influenza A virus and/or LPS in endothelial cells. The results obtained in the present work may be of use when studying the mechanisms of development of the influenza A virus and secondary bacterial infection complications.
Collapse
Affiliation(s)
- S S Smirnova
- 1Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia
| | - M M Pisareva
- 2Smorodintsev Research Institute of Influenza, Ministry of Healthcare of the Russian Federation, 197376 St. Petersburg, Russia
| | - T D Smirnova
- 2Smorodintsev Research Institute of Influenza, Ministry of Healthcare of the Russian Federation, 197376 St. Petersburg, Russia
| | - K V Sivak
- 2Smorodintsev Research Institute of Influenza, Ministry of Healthcare of the Russian Federation, 197376 St. Petersburg, Russia
| | - K V Vorobiev
- 1Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia
| |
Collapse
|