1
|
Shlapa Y, Siposova K, Sarnatskaya V, Drajnova M, Silvestre-Albero J, Lykhova O, Maraloiu VA, Solopan SO, Molcan M, Musatov A, Belous A. Bioactive Carbon@CeO 2 Composites as Efficient Antioxidants with Antiamyloid and Radioprotective Potentials. ACS APPLIED BIO MATERIALS 2024; 7:6749-6767. [PMID: 39320157 DOI: 10.1021/acsabm.4c00912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
Blending carbon particles (CPs) and nanoscale bioactive cerium dioxide is a promising approach for designing composites for biomedical applications, combining the sorption and antioxidant potentials of each individual component. To address this issue, it is crucial to assess the correlation between the components' ratio, physicochemical parameters, and biofunctionality of the composites. Thus, the current research was aimed at fabricating C@CeO2 composites with different molar ratios and the examination of how the parameters of the composites affect their bioactivity. XRD, X-ray photoelectron spectroscopy, and electron microscopy data verified the formation of C@CeO2 composites. CeO2 nanoparticles (NPs) of 4-6 nm are highly dispersed on the surfaces of amorphous CPs. The presence of CeO2 NPs on the carbon surface decreased its adsorption potential in a dose-dependent manner. Besides, the coexistence of carbon and CeO2 in a single composite promotes some redox interactions between O-functionalities and Ce3+/Ce4+ species, resulting in changes in the chemical state of the surface of the composites. These observations suggest the strong connection between these parameters and the biofunctionality of the composites. The presence of CeO2 NPs on the surface of carbon led to a significant increase in the stability of the prepared composites in their aqueous suspensions. The enhancement of bioactivity of the newly prepared C@CeO2 compared to bare carbon and CeO2 was validated by testing their pseudomimetic (catalase/peroxidase-like and superoxide dismutase-like), antiamyloid, and radioprotective activities.
Collapse
Affiliation(s)
- Yuliia Shlapa
- V. I. Vernadsky Institute of General & Inorganic Chemistry of the NAS of Ukraine, 32/34 Palladina Avenue, Kyiv 03142, Ukraine
| | - Katarina Siposova
- Institute of Experimental Physics, Slovak Academy of Science, Watsonova 47, Kosice 040 01, Slovakia
| | - Veronika Sarnatskaya
- R. E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology of the NAS of Ukraine, 45, Vasylkivska Street, Kyiv 03022, Ukraine
| | - Michaela Drajnova
- Institute of Experimental Physics, Slovak Academy of Science, Watsonova 47, Kosice 040 01, Slovakia
- Institute of Chemistry, Faculty of Science, P.J. Safarik University in Kosice, Moyzesova 11, Kosice 040 01, Slovakia
| | - Joaquin Silvestre-Albero
- Laboratorio de Materiales Avanzados, Instituto Universitario de Materiales-Departamento de Química Inorgánica, University of Alicante, Ctra. San Vicente-Alicante s/n, Alicante E-03080, Spain
| | - Olexandra Lykhova
- R. E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology of the NAS of Ukraine, 45, Vasylkivska Street, Kyiv 03022, Ukraine
| | | | - Sergii Oleksandrovich Solopan
- V. I. Vernadsky Institute of General & Inorganic Chemistry of the NAS of Ukraine, 32/34 Palladina Avenue, Kyiv 03142, Ukraine
| | - Matus Molcan
- Institute of Experimental Physics, Slovak Academy of Science, Watsonova 47, Kosice 040 01, Slovakia
| | - Andrey Musatov
- Institute of Experimental Physics, Slovak Academy of Science, Watsonova 47, Kosice 040 01, Slovakia
| | - Anatolii Belous
- V. I. Vernadsky Institute of General & Inorganic Chemistry of the NAS of Ukraine, 32/34 Palladina Avenue, Kyiv 03142, Ukraine
| |
Collapse
|
2
|
Zhang Q, Yaoita N, Tabuchi A, Liu S, Chen SH, Li Q, Hegemann N, Li C, Rodor J, Timm S, Laban H, Finkel T, Stevens T, Alvarez DF, Erfinanda L, de Perrot M, Kucherenko MM, Knosalla C, Ochs M, Dimmeler S, Korff T, Verma S, Baker AH, Kuebler WM. Endothelial Heterogeneity in the Response to Autophagy Drives Small Vessel Muscularization in Pulmonary Hypertension. Circulation 2024; 150:466-487. [PMID: 38873770 DOI: 10.1161/circulationaha.124.068726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 04/18/2024] [Indexed: 06/15/2024]
Abstract
BACKGROUND Endothelial cell (EC) apoptosis and proliferation of apoptosis-resistant cells is a hallmark of pulmonary hypertension (PH). Yet, why some ECs die and others proliferate and how this contributes to vascular remodeling is unclear. We hypothesized that this differential response may: (1) relate to different EC subsets, namely pulmonary artery (PAECs) versus microvascular ECs (MVECs); (2) be attributable to autophagic activation in both EC subtypes; and (3) cause replacement of MVECs by PAECs with subsequent distal vessel muscularization. METHODS EC subset responses to chronic hypoxia were assessed by single-cell RNA sequencing of murine lungs. Proliferative versus apoptotic responses, activation, and role of autophagy were assessed in human and rat PAECs and MVECs, and in precision-cut lung slices of wild-type mice or mice with endothelial deficiency in the autophagy-related gene 7 (Atg7EN-KO). Abundance of PAECs versus MVECs in precapillary microvessels was assessed in lung tissue from patients with PH and animal models on the basis of structural or surface markers. RESULTS In vitro and in vivo, PAECs proliferated in response to hypoxia, whereas MVECs underwent apoptosis. Single-cell RNA sequencing analyses support these findings in that hypoxia induced an antiapoptotic, proliferative phenotype in arterial ECs, whereas capillary ECs showed a propensity for cell death. These distinct responses were prevented in hypoxic Atg7EN-KO mice or after ATG7 silencing, yet replicated by autophagy stimulation. In lung tissue from mice, rats, or patients with PH, the abundance of PAECs in precapillary arterioles was increased, and that of MVECs reduced relative to controls, indicating replacement of microvascular by macrovascular ECs. EC replacement was prevented by genetic or pharmacological inhibition of autophagy in vivo. Conditioned medium from hypoxic PAECs yet not MVECs promoted pulmonary artery smooth muscle cell proliferation and migration in a platelet-derived growth factor-dependent manner. Autophagy inhibition attenuated PH development and distal vessel muscularization in preclinical models. CONCLUSIONS Autophagic activation by hypoxia induces in parallel PAEC proliferation and MVEC apoptosis. These differential responses cause a progressive replacement of MVECs by PAECs in precapillary pulmonary arterioles, thus providing a macrovascular context that in turn promotes pulmonary artery smooth muscle cell proliferation and migration, ultimately driving distal vessel muscularization and the development of PH.
Collapse
Affiliation(s)
- Qi Zhang
- Institute of Physiology (Q.Z., N.Y., A.T., S.L., Q.L., N.H., C.L., L.E., M.M.K., W.M.K.), Charité-Universitätsmedizin, Berlin, Germany
- Department of Cardiology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China (Q.Z.)
| | - Nobuhiro Yaoita
- Institute of Physiology (Q.Z., N.Y., A.T., S.L., Q.L., N.H., C.L., L.E., M.M.K., W.M.K.), Charité-Universitätsmedizin, Berlin, Germany
| | - Arata Tabuchi
- Institute of Physiology (Q.Z., N.Y., A.T., S.L., Q.L., N.H., C.L., L.E., M.M.K., W.M.K.), Charité-Universitätsmedizin, Berlin, Germany
| | - Shaofei Liu
- Institute of Physiology (Q.Z., N.Y., A.T., S.L., Q.L., N.H., C.L., L.E., M.M.K., W.M.K.), Charité-Universitätsmedizin, Berlin, Germany
- German Center for Cardiovascular Research, Partner Site Berlin (S.L., N.H., M.M.K., C.K., W.M.K.)
| | - Shiau-Haln Chen
- Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (S.-H.C., J.R., A.H.B.)
| | - Qiuhua Li
- Institute of Physiology (Q.Z., N.Y., A.T., S.L., Q.L., N.H., C.L., L.E., M.M.K., W.M.K.), Charité-Universitätsmedizin, Berlin, Germany
| | - Niklas Hegemann
- Institute of Physiology (Q.Z., N.Y., A.T., S.L., Q.L., N.H., C.L., L.E., M.M.K., W.M.K.), Charité-Universitätsmedizin, Berlin, Germany
- German Center for Cardiovascular Research, Partner Site Berlin (S.L., N.H., M.M.K., C.K., W.M.K.)
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité, Berlin, Germany (N.H., M.M.K., C.K.)
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Germany (N.H., M.M.K., C.K.)
| | - Caihong Li
- Institute of Physiology (Q.Z., N.Y., A.T., S.L., Q.L., N.H., C.L., L.E., M.M.K., W.M.K.), Charité-Universitätsmedizin, Berlin, Germany
| | - Julie Rodor
- Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (S.-H.C., J.R., A.H.B.)
| | - Sara Timm
- Core Facility Electron Microscopy (S.T., M.O.), Charité-Universitätsmedizin, Berlin, Germany
| | - Hebatullah Laban
- Institute of Physiology and Pathophysiology, Department of Cardiovascular Physiology (H.L.), Heidelberg University, Germany
- German Center for Cardiovascular Research, Partner Site Heidelberg/Mannheim, Heidelberg (H.L.)
| | - Toren Finkel
- Department of Medicine, Division of Cardiology, University of Pittsburgh, PA (T.F.)
| | - Troy Stevens
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile (T.S.)
| | - Diego F Alvarez
- Department of Physiology and Pharmacology, College of Osteopathic Medicine, Sam Houston State University, Conroe, TX (D.F.A.)
| | - Lasti Erfinanda
- Institute of Physiology (Q.Z., N.Y., A.T., S.L., Q.L., N.H., C.L., L.E., M.M.K., W.M.K.), Charité-Universitätsmedizin, Berlin, Germany
| | - Marc de Perrot
- Division of Thoracic Surgery, Toronto General Hospital, Canada (M.d.P.)
- Department of Surgery (M.d.P., W.M.K.), University of Toronto, Canada
| | - Mariya M Kucherenko
- Institute of Physiology (Q.Z., N.Y., A.T., S.L., Q.L., N.H., C.L., L.E., M.M.K., W.M.K.), Charité-Universitätsmedizin, Berlin, Germany
- German Center for Cardiovascular Research, Partner Site Berlin (S.L., N.H., M.M.K., C.K., W.M.K.)
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité, Berlin, Germany (N.H., M.M.K., C.K.)
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Germany (N.H., M.M.K., C.K.)
| | - Christoph Knosalla
- German Center for Cardiovascular Research, Partner Site Berlin (S.L., N.H., M.M.K., C.K., W.M.K.)
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité, Berlin, Germany (N.H., M.M.K., C.K.)
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Germany (N.H., M.M.K., C.K.)
| | - Matthias Ochs
- Core Facility Electron Microscopy (S.T., M.O.), Charité-Universitätsmedizin, Berlin, Germany
- Institute of Functional Anatomy (M.O.), Charité-Universitätsmedizin, Berlin, Germany
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany (S.D.)
| | - Thomas Korff
- Department of Cardiovascular Physiology, Institute of Physiology and Pathophysiology (T.K.), Heidelberg University, Germany
- European Center for Angioscience, Medical Faculty Mannheim (T.K.), Heidelberg University, Germany
| | - Subodh Verma
- Division of Cardiac Surgery (S.V.), University of Toronto, Canada
| | - Andrew H Baker
- Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (S.-H.C., J.R., A.H.B.)
- Department of Pathology, Cardiovascular Research Institute Maastricht School for Cardiovascular Diseases, Maastricht University, The Netherlands (A.H.B.)
| | - Wolfgang M Kuebler
- Institute of Physiology (Q.Z., N.Y., A.T., S.L., Q.L., N.H., C.L., L.E., M.M.K., W.M.K.), Charité-Universitätsmedizin, Berlin, Germany
- German Center for Cardiovascular Research, Partner Site Berlin (S.L., N.H., M.M.K., C.K., W.M.K.)
- Department of Surgery (M.d.P., W.M.K.), University of Toronto, Canada
- Department of Physiology (W.M.K.), University of Toronto, Canada
- Keenan Research Centre, St Michael's Hospital, Canada (W.M.K.)
| |
Collapse
|
3
|
Zhang C, Guo ZF, Liu W, Kazama K, Hu L, Sun X, Wang L, Lee H, Lu L, Yang XF, Summer R, Sun J. PIMT is a novel and potent suppressor of endothelial activation. eLife 2023; 12:e85754. [PMID: 37070640 PMCID: PMC10112892 DOI: 10.7554/elife.85754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 04/02/2023] [Indexed: 04/19/2023] Open
Abstract
Proinflammatory agonists provoke the expression of cell surface adhesion molecules on endothelium in order to facilitate leukocyte infiltration into tissues. Rigorous control over this process is important to prevent unwanted inflammation and organ damage. Protein L-isoaspartyl O-methyltransferase (PIMT) converts isoaspartyl residues to conventional methylated forms in cells undergoing stress-induced protein damage. The purpose of this study was to determine the role of PIMT in vascular homeostasis. PIMT is abundantly expressed in mouse lung endothelium and PIMT deficiency in mice exacerbated pulmonary inflammation and vascular leakage to LPS(lipopolysaccharide). Furthermore, we found that PIMT inhibited LPS-induced toll-like receptor signaling through its interaction with TNF receptor-associated factor 6 (TRAF6) and its ability to methylate asparagine residues in the coiled-coil domain. This interaction was found to inhibit TRAF6 oligomerization and autoubiquitination, which prevented NF-κB transactivation and subsequent expression of endothelial adhesion molecules. Separately, PIMT also suppressed ICAM-1 expression by inhibiting its N-glycosylation, causing effects on protein stability that ultimately translated into reduced EC(endothelial cell)-leukocyte interactions. Our study has identified PIMT as a novel and potent suppressor of endothelial activation. Taken together, these findings suggest that therapeutic targeting of PIMT may be effective in limiting organ injury in inflammatory vascular diseases.
Collapse
Affiliation(s)
- Chen Zhang
- Center for Translational Medicine, Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Zhi-Fu Guo
- Center for Translational Medicine, Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Wennan Liu
- Center for Translational Medicine, Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Kyosuke Kazama
- Center for Translational Medicine, Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Louis Hu
- Center for Translational Medicine, Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Xiaobo Sun
- Center for Translational Medicine, Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Lu Wang
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Hyoungjoo Lee
- Quantitative Proteomics Resource Center, University of PennsylvaniaPhiladelphiaUnited States
| | - Lin Lu
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xiao-Feng Yang
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple UniversityPhiladelphiaUnited States
| | - Ross Summer
- Center for Translational Medicine, Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Jianxin Sun
- Center for Translational Medicine, Thomas Jefferson UniversityPhiladelphiaUnited States
| |
Collapse
|
4
|
Turova V, Kovtanyuk A, Pykhteev O, Sidorenko I, Lampe R. Glycocalyx Sensing with a Mathematical Model of Acoustic Shear Wave Biosensor. Bioengineering (Basel) 2022; 9:462. [PMID: 36135008 PMCID: PMC9495919 DOI: 10.3390/bioengineering9090462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/12/2022] [Accepted: 08/22/2022] [Indexed: 11/22/2022] Open
Abstract
The article deals with an idea of exploiting an acoustic shear wave biosensor for investigating the glycocalyx, a polysaccharide polymer molecule layer on the endothelium of blood vessels that, according to recent studies, plays an important role in protecting against diseases. To test this idea, a mathematical model of an acoustic shear wave sensor and corresponding software developed earlier for proteomic applications are used. In this case, the glycocalyx is treated as a layer homogenized over the thin polymer "villi". Its material characteristics depend on the density, thickness, and length of the villi and on the viscous properties of the surrounding liquid (blood plasma). It is proved that the model used has a good sensitivity to the above parameters of the villi and blood plasma. Numerical experiments performed using real data collected retrospectively from premature infants show that the use of acoustic shear wave sensors may be a promising approach to investigate properties of glycocalyx-like structures and their role in prematurity.
Collapse
Affiliation(s)
- Varvara Turova
- Klinikum Rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675 München, Germany
| | - Andrey Kovtanyuk
- Klinikum Rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675 München, Germany
- Fakultät für Mathematik, Technische Universität München, Boltzmannstr. 3, 85747 Garching bei München, Germany
| | | | - Irina Sidorenko
- Fakultät für Mathematik, Technische Universität München, Boltzmannstr. 3, 85747 Garching bei München, Germany
| | - Renée Lampe
- Klinikum Rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675 München, Germany
| |
Collapse
|
5
|
Zhao F, Satyanarayana G, Zhang Z, Zhao J, Ma XL, Wang Y. Endothelial Autophagy in Coronary Microvascular Dysfunction and Cardiovascular Disease. Cells 2022; 11:2081. [PMID: 35805165 PMCID: PMC9265562 DOI: 10.3390/cells11132081] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/22/2022] [Accepted: 06/28/2022] [Indexed: 02/06/2023] Open
Abstract
Coronary microvascular dysfunction (CMD) refers to a subset of structural and/or functional disorders of coronary microcirculation that lead to impaired coronary blood flow and eventually myocardial ischemia. Amid the growing knowledge of the pathophysiological mechanisms and the development of advanced tools for assessment, CMD has emerged as a prevalent cause of a broad spectrum of cardiovascular diseases (CVDs), including obstructive and nonobstructive coronary artery disease, diabetic cardiomyopathy, and heart failure with preserved ejection fraction. Of note, the endothelium exerts vital functions in regulating coronary microvascular and cardiac function. Importantly, insufficient or uncontrolled activation of endothelial autophagy facilitates the pathogenesis of CMD in diverse CVDs. Here, we review the progress in understanding the pathophysiological mechanisms of autophagy in coronary endothelial cells and discuss their potential role in CMD and CVDs.
Collapse
Affiliation(s)
- Fujie Zhao
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA; (F.Z.); (Z.Z.); (J.Z.); (X.-L.M.)
| | | | - Zheng Zhang
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA; (F.Z.); (Z.Z.); (J.Z.); (X.-L.M.)
| | - Jianli Zhao
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA; (F.Z.); (Z.Z.); (J.Z.); (X.-L.M.)
| | - Xin-Liang Ma
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA; (F.Z.); (Z.Z.); (J.Z.); (X.-L.M.)
| | - Yajing Wang
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA; (F.Z.); (Z.Z.); (J.Z.); (X.-L.M.)
| |
Collapse
|
6
|
Subsets of Cytokines and Chemokines from DENV-4-Infected Patients Could Regulate the Endothelial Integrity of Cultured Microvascular Endothelial Cells. Pathogens 2022; 11:pathogens11050509. [PMID: 35631030 PMCID: PMC9144803 DOI: 10.3390/pathogens11050509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/14/2022] [Accepted: 04/19/2022] [Indexed: 11/17/2022] Open
Abstract
Introduction: It is a consensus that inflammatory mediators produced by immune cells contribute to changes in endothelial permeability in dengue. We propose to relate inflammatory mediators seen in dengue patients with the in vitro alteration of endothelial cells (ECs) cultured with serum from these patients. Methods: Patients with mild (DF) to moderate and severe dengue (DFWS/Sev) were selected. ELISA quantified inflammatory mediators. Expression of adhesion molecules and CD147 were evaluated in the ECs cultured with the patient’s serum by flow cytometry. We assessed endothelial permeability by measuring transendothelial electrical resistance in cocultures of ECs with patient serum. Results: Dengue infection led to an increase in inflammatory mediators—the IL-10 distinguished DF from DFWS/Sev. There were no changes in CD31, CD54, and CD106 but decreased CD147 expression in ECs. DFWS/Sev sera induced a greater difference in endothelial permeability than DF sera. Correlation statistical test indicated that low IL-10 and IFN-γ and high CCL5 maintain the integrity of ECs in DF patients. In contrast, increased TNF, IFN-γ, CXCL8, and CCL2 maintain EC integrity in DFWS/Sev patients. Conclusions: Our preliminary data suggest that a subset of inflammatory mediators may be related to the maintenance or loss of endothelial integrity, reflecting the clinical prognosis.
Collapse
|
7
|
Silva M, Coelho A, Vargas S, Faustino P. VCAM1, HMOX1 and NOS3 differential endothelial expression may impact sickle cell anemia vasculopathy. Blood Cells Mol Dis 2022; 93:102639. [PMID: 34999313 DOI: 10.1016/j.bcmd.2021.102639] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 12/21/2021] [Accepted: 12/21/2021] [Indexed: 10/19/2022]
Abstract
Endothelial dysfunction plays a major role in sickle cell anemia (SCA) systemic vasculopathy, with upregulation of adhesion molecules (e.g., VCAM-1), decreased nitric oxide bioavailability, and oxidative stress. We aimed to assess the modulation role of pro-inflammatory and pro-oxidative stimuli on endothelial VCAM1, NOS3, and HMOX1 expression. We also evaluated the effect of the main SCA therapeutic agent, hydroxyurea, on that modulation. Our results showed that two VCAM1 promoter haplotypes, we previously associated with pediatric cerebral vasculopathy and severe hemolysis in SCA, increased promoter activity in TNF-α-stimulated transfected EA.hy926 and HBEC cell lines, consistent with a higher VCAM1 expression in macro and microvascular settings. In non-transfected cells, we also observed TNF-α-induced VCAM1 overexpression as well as heme-induced overexpression of HMOX1 in both cell models. Heme did not affect VCAM1 nor NOS3 expression and the latter was also not affected by TNF-α stimulus. Hydroxyurea treatment lowered TNF-induced VCAM1 and NOS3 expression but did not affect heme-induced HMOX1 expression. These data further indicate that VCAM1 haplotypes we studied lead to higher VCAM1 expression affecting not only cerebral but also systemic vasculopathy risk. The differential endothelial expression of VCAM1, NOS3, and HMOX1 also confirms their genetic modulation role in SCA systemic vasculopathy.
Collapse
Affiliation(s)
- Marisa Silva
- Departamento de Genética Humana, Instituto Nacional de Saúde Dr. Ricardo Jorge, Lisbon, Portugal
| | - Andreia Coelho
- Departamento de Genética Humana, Instituto Nacional de Saúde Dr. Ricardo Jorge, Lisbon, Portugal
| | - Sofia Vargas
- Departamento de Genética Humana, Instituto Nacional de Saúde Dr. Ricardo Jorge, Lisbon, Portugal
| | - Paula Faustino
- Departamento de Genética Humana, Instituto Nacional de Saúde Dr. Ricardo Jorge, Lisbon, Portugal; Instituto de Saúde Ambiental (ISAMB), Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal; Laboratório Associado TERRA, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
8
|
Tarnawski AS, Ahluwalia A. Endothelial cells and blood vessels are major targets for COVID-19-induced tissue injury and spreading to various organs. World J Gastroenterol 2022; 28:275-289. [PMID: 35110950 PMCID: PMC8771611 DOI: 10.3748/wjg.v28.i3.275] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/02/2021] [Accepted: 01/11/2022] [Indexed: 02/06/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) infected so far over 250 million people and caused the death of over 5 million worldwide. Aging, diabetes, and cardiovascular diseases, conditions with preexisting impaired endothelial functions predispose to COVID-19. While respiratory epithelium is the main route of virus entry, the endothelial cells (ECs) lining pulmonary blood vessels are also an integral part of lung injury in COVID-19 patients. COVID-19 not only affects the lungs and respiratory system but also gastrointestinal (GI) tract, liver, pancreas, kidneys, heart, brain, and skin. Blood vessels are likely conduits for the virus dissemination to these distant organs. Importantly, ECs are also critical for vascular regeneration during injury/lesions healing and restoration of vascular network. The World Journal of Gastroenterology has published in last two years over 67 outstanding papers on COVID-19 infection with a focus on the GI tract, liver, pancreas, etc., however, the role of the endothelial and vascular components as major targets for COVID-19-induced tissue injury, spreading to various organs, and injury healing have not been sufficiently emphasized. In the present article, we focus on these subjects and on current treatments including the most recent oral drugs molnupiravir and paxlovid that show a dramatic, significant efficacy in controlling severe COVID-19 infection.
Collapse
Affiliation(s)
- Andrzej S Tarnawski
- Gastroenterology Research Department, University of California Irvine and the Veterans Administration Long Beach Healthcare System, Long Beach, CA 90822, United States
| | - Amrita Ahluwalia
- Research Service, Veterans Administration Long Beach Healthcare System, Long Beach, CA 90822, United States
| |
Collapse
|
9
|
Pan CS, Yan L, Lin SQ, He K, Cui YC, Liu YY, Hu BH, Chang X, Zhao XR, Fan JY, Han JY. QiShenYiQi Pills Attenuates Ischemia/Reperfusion-Induced Cardiac Microvascular Hyperpermeability Implicating Src/Caveolin-1 and RhoA/ROCK/MLC Signaling. Front Physiol 2022; 12:753761. [PMID: 34975519 PMCID: PMC8718710 DOI: 10.3389/fphys.2021.753761] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 11/18/2021] [Indexed: 01/25/2023] Open
Abstract
Aims: Coronary microvascular hyperpermeability is an important contributor to ischemia or reperfusion (I/R) injury. However, the effective strategy for this insult remains limited. This study aimed to explore the protective effect of the compound Chinese medicine QiShenYiQi Pills (QSYQ) against coronary microvascular hyperpermeability after cardiac I/R with focusing on the underlying mechanism. Methods and Results: Male Sprague-Dawley rats under anesthesia were subjected to occlusion of left coronary anterior descending artery followed by reperfusion. QSYQ was administrated 90 min before ischemia initiation. Human cardiac microvascular endothelial cells (HCMECs) underwent hypoxia or reoxygenation (H/R) challenge with QSYQ administrated 1 h prior to hypoxia. QSYQ exhibited effects on attenuating microvascular damage and albumin leakage after I/R injury, showing a role in maintaining endothelial junctions, caveolae, and collagen in basement membrane (BM) of microvessels. Study using HCMECs disclosed that QSYQ protected endothelial barrier from impairment by H/R, attenuating the decline of respiratory chain complex I and ATP synthase, activation of Src/caveolin-1 and increase of RhoA/ROCK/p-MLC, MMP-9, and CTSS. PP2, a Src inhibitor, partially imitated the effect of QSYQ. Conclusions: The QSYQ was able to prevent I/R-induced cardiac microvascular hyperpermeability via a mechanism involving Src/caveolin-1 and RhoA/ROCK/MLC signaling.
Collapse
Affiliation(s)
- Chun-Shui Pan
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Li Yan
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Se-Qi Lin
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Ke He
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yuan-Chen Cui
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yu-Ying Liu
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Bai-He Hu
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Xin Chang
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Xin-Rong Zhao
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jing-Yu Fan
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jing-Yan Han
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China.,State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tianjin, China
| |
Collapse
|
10
|
Animal Models of Neointimal Hyperplasia and Restenosis: Species-Specific Differences and Implications for Translational Research. JACC Basic Transl Sci 2021; 6:900-917. [PMID: 34869956 PMCID: PMC8617545 DOI: 10.1016/j.jacbts.2021.06.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 06/17/2021] [Accepted: 06/20/2021] [Indexed: 12/29/2022]
Abstract
Neointimal hyperplasia is the major factor contributing to restenosis after angioplasty procedures. Multiple animal models exist to study basic and translational aspects of restenosis formation. Animal models differ substantially, and species-specific differences have major impact on the pathophysiology of the model. Genetic, dietary, and mechanical interventions determine the translational potential of the animal model used and have to be considered when choosing the model.
The process of restenosis is based on the interplay of various mechanical and biological processes triggered by angioplasty-induced vascular trauma. Early arterial recoil, negative vascular remodeling, and neointimal formation therefore limit the long-term patency of interventional recanalization procedures. The most serious of these processes is neointimal hyperplasia, which can be traced back to 4 main mechanisms: endothelial damage and activation; monocyte accumulation in the subintimal space; fibroblast migration; and the transformation of vascular smooth muscle cells. A wide variety of animal models exists to investigate the underlying pathophysiology. Although mouse models, with their ease of genetic manipulation, enable cell- and molecular-focused fundamental research, and rats provide the opportunity to use stent and balloon models with high throughput, both rodents lack a lipid metabolism comparable to humans. Rabbits instead build a bridge to close the gap between basic and clinical research due to their human-like lipid metabolism, as well as their size being accessible for clinical angioplasty procedures. Every different combination of animal, dietary, and injury model has various advantages and disadvantages, and the decision for a proper model requires awareness of species-specific biological properties reaching from vessel morphology to distinct cellular and molecular features.
Collapse
Key Words
- Apo, apolipoprotein
- CETP, cholesteryl ester transferase protein
- ECM, extracellular matrix
- FGF, fibroblast growth factor
- HDL, high-density lipoprotein
- LDL, low-density lipoprotein
- LDLr, LDL receptor
- PDGF, platelet-derived growth factor
- TGF, transforming growth factor
- VLDL, very low-density lipoprotein
- VSMC, vascular smooth muscle cell
- angioplasty
- animal model
- neointimal hyperplasia
- restenosis
Collapse
|
11
|
Fu S, Yu S, Zhao Y, Ma X, Li X. Unfractionated Heparin Attenuated Histone-Induced Pulmonary Syndecan-1 Degradation in Mice: a Preliminary Study on the Roles of Heparinase Pathway. Inflammation 2021; 45:712-724. [PMID: 34657233 DOI: 10.1007/s10753-021-01578-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/07/2021] [Accepted: 10/05/2021] [Indexed: 10/20/2022]
Abstract
Endothelial glycocalyx degradation is thought to facilitate the development of sepsis. Histone is a significant mediator in sepsis. Unfractionated heparin (UFH) possessed beneficial effects on sepsis. Thereby, this study aims to figure out whether histone can disrupt glycocalyx and to investigate the protective effect and mechanism of UFH. Male mice (C57BL/6, 8-10 weeks old, weighing 20-25 g) were randomly divided into five groups including control group, histone group, histone + UFH group, histone + heparinase (HPA) inhibitor group, and histone + UFH + HPA inhibitor group. The mice were treated with histone (50 mg/kg) via tail vein immediately after HPA (20 mg/kg) injection. UFH (400 U/kg) was injected 1h after histone administration. The other groups were injected with equal volume of sterile saline accordingly. UFH alleviated histone-induced lung injury and pulmonary edema. UFH inhibited histone-induced lung coagulation activation and inflammatory response. UFH treatment markedly inhibited pulmonary glycocalyx degradation by reducing the histone-induced decrease in the levels of lung syndecan-1 mRNA and protein. UFH downregulated histone-induced expression of HPA mRNA and protein, and thus alleviated glycocalyx degradation. UFH protects against histone-induced pulmonary glycocalyx injury partly by heparinase pathway.
Collapse
Affiliation(s)
- Sifeng Fu
- Department of Critical Care Medicine, the First Affiliated Hospital, China Medical University, North Nanjing Street 155, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Sihan Yu
- Department of Critical Care Medicine, the First Affiliated Hospital, China Medical University, North Nanjing Street 155, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Yilin Zhao
- Department of Critical Care Medicine, the First Affiliated Hospital, China Medical University, North Nanjing Street 155, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Xiaochun Ma
- Department of Critical Care Medicine, the First Affiliated Hospital, China Medical University, North Nanjing Street 155, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Xu Li
- Department of Critical Care Medicine, the First Affiliated Hospital, China Medical University, North Nanjing Street 155, Shenyang, 110001, Liaoning Province, People's Republic of China.
| |
Collapse
|
12
|
Hulde N, Rogenhofer N, Brettner F, Eckert NC, Fetz I, Buchheim JI, Kammerer T, Dendorfer A, Choukèr A, Hofmann-Kiefer KF, Rehm M, Thaler C. Effects of controlled ovarian stimulation on vascular barrier and endothelial glycocalyx: a pilot study. J Assist Reprod Genet 2021; 38:2273-2282. [PMID: 34286421 PMCID: PMC8490540 DOI: 10.1007/s10815-021-02233-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 05/11/2021] [Indexed: 11/26/2022] Open
Abstract
Purpose Controlled ovarian stimulation significantly amplifies the number of maturing and ovulated follicles as well as ovarian steroid production. The ovarian hyperstimulation syndrome (OHSS) increases capillary permeability and fluid extravasation. Vascular integrity intensely is regulated by an endothelial glycocalyx (EGX) and we have shown that ovulatory cycles are associated with shedding of EGX components. This study investigates if controlled ovarian stimulation impacts on the integrity of the endothelial glycocalyx as this might explain key pathomechanisms of the OHSS. Methods Serum levels of endothelial glycocalyx components of infertility patients (n=18) undergoing controlled ovarian stimulation were compared to a control group of healthy women with regular ovulatory cycles (n=17). Results Patients during luteal phases of controlled ovarian stimulation cycles as compared to normal ovulatory cycles showed significantly increased Syndecan-1 serum concentrations (12.6 ng/ml 6.1125th–19.1375th to 13.9 ng/ml 9.625th–28.975th; p=0.026), indicating shedding and degradation of the EGX. Conclusion A shedding of EGX components during ovarian stimulation has not yet been described. Our study suggests that ovarian stimulation may affect the integrity of the endothelial surface layer and increasing vascular permeability. This could explain key features of the OHSS and provide new ways of prevention of this serious condition of assisted reproduction.
Collapse
Affiliation(s)
- Nikolai Hulde
- Department of Anesthesiology, Heart and Diabetes Center North Rhine-Westphalia, Ruhr University Bochum, Georgstr 11, 32545, Bad Oeynhausen, Germany.
| | - N Rogenhofer
- Division of Gynecological Endocrinology and Reproductive Medicine, Department of Gynecology and Obstetrics, Ludwig-Maximilians University, Marchioninistr. 15, 81377, Munich, Germany
| | - F Brettner
- Department of Anesthesiology, Ludwig-Maximilians University, Marchioninistr. 15, 81377, Munich, Germany
| | - N C Eckert
- Department of Anesthesiology, Ludwig-Maximilians University, Marchioninistr. 15, 81377, Munich, Germany
| | - I Fetz
- Department of Anesthesiology, Ludwig-Maximilians University, Marchioninistr. 15, 81377, Munich, Germany
| | - J-I Buchheim
- Department of Anesthesiology, Ludwig-Maximilians University, Marchioninistr. 15, 81377, Munich, Germany
| | - T Kammerer
- Department of Anesthesiology, Ludwig-Maximilians University, Marchioninistr. 15, 81377, Munich, Germany
| | - A Dendorfer
- Walter-Brendel-Centre of Experimental Medicine, Hospital of the University Munich, Ludwig-Maximilians University, Marchioninistr. 15, 81377, Munich, Germany
| | - A Choukèr
- Department of Anesthesiology, Ludwig-Maximilians University, Marchioninistr. 15, 81377, Munich, Germany
| | - K F Hofmann-Kiefer
- Department of Anesthesiology, Ludwig-Maximilians University, Marchioninistr. 15, 81377, Munich, Germany
| | - M Rehm
- Department of Anesthesiology, Ludwig-Maximilians University, Marchioninistr. 15, 81377, Munich, Germany
| | - C Thaler
- Division of Gynecological Endocrinology and Reproductive Medicine, Department of Gynecology and Obstetrics, Ludwig-Maximilians University, Marchioninistr. 15, 81377, Munich, Germany
| |
Collapse
|
13
|
Yokomori H, Ando W, Oda M. Plasmalemmal Vesicle-Associated Protein Is Associated with Endothelial Cells Sprouting from the Peribiliary Capillary Plexus in Human Cirrhotic Liver. J Vasc Res 2021; 58:361-369. [PMID: 34280928 DOI: 10.1159/000516923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 04/19/2021] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Plasmalemmal vesicle-associated protein (PLVAP) is an endothelial-specific integral membrane glycoprotein that localizes to caveolae and fenestrae in animal models; however, little is known about PLVAP in endothelial cells (ECs) in hepatic sinusoids during liver cirrhosis (LC). Here, we aimed to elucidate PLVAP localization and expression in the human liver during LC progression. METHODS PLVAP protein expression was detected in specimens from normal control livers and hepatitis C-related cirrhotic livers using immunohistochemistry, Western blotting, and immunoelectron microscopy. RESULTS PLVAP mainly localized to the peribiliary capillary plexus (PCP) and was rarely observed in hepatic artery branches and portal venules in control tissue, but was aberrantly expressed in capillarized sinusoids and proliferated capillaries in fibrotic septa within cirrhotic liver tissue. Ultrastructural analysis indicated that PLVAP localized to thin ECs in some caveolae, whereas PLVAP localized primarily to caveolae-like structures and proliferative sinusoid capillary EC vesicles in cirrhotic liver tissue. Western blot analysis confirmed that PLVAP was overexpressed at the protein level in advanced cirrhotic liver tissue. CONCLUSION PLVAP was strongly expressed in the caveolae of proliferated capillaries directly connected with sinusoids linked with the PCP, suggesting that it plays a role in angiogenesis and sinusoidal remodeling in LC.
Collapse
Affiliation(s)
- Hiroaki Yokomori
- Department of Internal Medicine, Kitasato University Medical Center, Saitama, Japan
| | - Wataru Ando
- Department of Pharmaceutical Science, Kitasato University, Tokyo, Japan
| | - Masaya Oda
- The Chunichi Newspapers, The Main Tokyo Clinic, Tokyo, Japan
| |
Collapse
|
14
|
Villalba N, Baby S, Yuan SY. The Endothelial Glycocalyx as a Double-Edged Sword in Microvascular Homeostasis and Pathogenesis. Front Cell Dev Biol 2021; 9:711003. [PMID: 34336864 PMCID: PMC8316827 DOI: 10.3389/fcell.2021.711003] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 06/22/2021] [Indexed: 12/27/2022] Open
Abstract
Expressed on the endothelial cell (EC) surface of blood vessels, the glycocalyx (GCX), a mixture of carbohydrates attached to proteins, regulates the access of cells and molecules in the blood to the endothelium. Besides protecting endothelial barrier integrity, the dynamic microstructure of the GCX confers remarkable functions including mechanotransduction and control of vascular tone. Recently, a novel perspective has emerged supporting the pleiotropic roles of the endothelial GCX (eGCX) in cardiovascular health and disease. Because eGCX degradation occurs in certain pathological states, the circulating levels of eGCX degradation products have been recognized to have diagnostic or prognostic values. Beyond their biomarker roles, certain eGCX fragments serve as pathogenic factors in disease progression. Pharmacological interventions that attenuate eGCX degradation or restore its integrity have been sought. This review provides our current understanding of eGCX structure and function across the microvasculature in different organs. We also discuss disease or injury states, such as infection, sepsis and trauma, where eGCX dysfunction contributes to severe inflammatory vasculopathy.
Collapse
Affiliation(s)
- Nuria Villalba
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Sheon Baby
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Sarah Y Yuan
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States.,Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| |
Collapse
|
15
|
Chen L, Holder R, Porter C, Shah Z. Vitamin D3 attenuates doxorubicin-induced senescence of human aortic endothelial cells by upregulation of IL-10 via the pAMPKα/Sirt1/Foxo3a signaling pathway. PLoS One 2021; 16:e0252816. [PMID: 34101754 PMCID: PMC8186764 DOI: 10.1371/journal.pone.0252816] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 05/21/2021] [Indexed: 01/10/2023] Open
Abstract
The toxicity of doxorubicin to the cardiovascular system often limits its benefits and widespread use as chemotherapy. The mechanisms involved in doxorubicin-induced cardiovascular damage and possible protective interventions are not well-explored. Using human aortic endothelial cells, we show vitamin D3 strongly attenuates doxorubicin-induced senescence and cell cycle arrest. We further show the protective effects of vitamin D3 are mediated by the upregulation of IL-10 and FOXO3a expression through fine modulation of pAMPKα/SIRT1/FOXO3a complex activity. These results have great significance in finding a target for mitigating doxorubicin-induced cardiovascular toxicity.
Collapse
Affiliation(s)
- Lei Chen
- Department of Cardiovascular Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Rachel Holder
- Department of Cardiovascular Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Charles Porter
- Department of Cardiovascular Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Zubair Shah
- Department of Cardiovascular Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
- * E-mail:
| |
Collapse
|
16
|
Narezkina A, Narayan HK, Zemljic-Harpf AE. Molecular mechanisms of anthracycline cardiovascular toxicity. Clin Sci (Lond) 2021; 135:1311-1332. [PMID: 34047339 PMCID: PMC10866014 DOI: 10.1042/cs20200301] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 05/07/2021] [Accepted: 05/11/2021] [Indexed: 12/21/2022]
Abstract
Anthracyclines are effective chemotherapeutic agents, commonly used in the treatment of a variety of hematologic malignancies and solid tumors. However, their use is associated with a significant risk of cardiovascular toxicities and may result in cardiomyopathy and heart failure. Cardiomyocyte toxicity occurs via multiple molecular mechanisms, including topoisomerase II-mediated DNA double-strand breaks and reactive oxygen species (ROS) formation via effects on the mitochondrial electron transport chain, NADPH oxidases (NOXs), and nitric oxide synthases (NOSs). Excess ROS may cause mitochondrial dysfunction, endoplasmic reticulum stress, calcium release, and DNA damage, which may result in cardiomyocyte dysfunction or cell death. These pathophysiologic mechanisms cause tissue-level manifestations, including characteristic histopathologic changes (myocyte vacuolization, myofibrillar loss, and cell death), atrophy and fibrosis, and organ-level manifestations including cardiac contractile dysfunction and vascular dysfunction. In addition, these mechanisms are relevant to current and emerging strategies to diagnose, prevent, and treat anthracycline-induced cardiomyopathy. This review details the established and emerging data regarding the molecular mechanisms of anthracycline-induced cardiovascular toxicity.
Collapse
Affiliation(s)
- Anna Narezkina
- Department of Medicine, Division of Cardiovascular Medicine, UCSD Cardiovascular Institute, University of California, San Diego
| | - Hari K. Narayan
- Department of Pediatrics, Division of Cardiology, University of California, San Diego
| | - Alice E. Zemljic-Harpf
- Veterans Affairs San Diego Healthcare System, San Diego, USA
- Department of Anesthesiology, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
17
|
The Endothelial Glycocalyx and Organ Preservation-From Physiology to Possible Clinical Implications for Solid Organ Transplantation. Int J Mol Sci 2021; 22:ijms22084019. [PMID: 33924713 PMCID: PMC8070558 DOI: 10.3390/ijms22084019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/09/2021] [Accepted: 04/12/2021] [Indexed: 01/14/2023] Open
Abstract
The endothelial glycocalyx is a thin layer consisting of proteoglycans, glycoproteins and glycosaminoglycans that lines the luminal side of vascular endothelial cells. It acts as a barrier and contributes to the maintenance of vascular homeostasis and microperfusion. During solid organ transplantation, the endothelial glycocalyx of the graft is damaged as part of Ischemia Reperfusion Injury (IRI), which is associated with impaired organ function. Although several substances are known to mitigate glycocalyx damage, it has not been possible to use these substances during graft storage on ice. Normothermic machine perfusion (NMP) emerges as an alternative technology for organ preservation and allows for organ evaluation, but also offers the possibility to treat and thus improve organ quality during storage. This review highlights the current knowledge on glycocalyx injury during organ transplantation, presents ways to protect the endothelial glycocalyx and discusses potential glycocalyx protection strategies during normothermic machine perfusion.
Collapse
|
18
|
Wolpe AG, Ruddiman CA, Hall PJ, Isakson BE. Polarized Proteins in Endothelium and Their Contribution to Function. J Vasc Res 2021; 58:65-91. [PMID: 33503620 DOI: 10.1159/000512618] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 10/27/2020] [Indexed: 12/11/2022] Open
Abstract
Protein localization in endothelial cells is tightly regulated to create distinct signaling domains within their tight spatial restrictions including luminal membranes, abluminal membranes, and interendothelial junctions, as well as caveolae and calcium signaling domains. Protein localization in endothelial cells is also determined in part by the vascular bed, with differences between arteries and veins and between large and small arteries. Specific protein polarity and localization is essential for endothelial cells in responding to various extracellular stimuli. In this review, we examine protein localization in the endothelium of resistance arteries, with occasional references to other vessels for contrast, and how that polarization contributes to endothelial function and ultimately whole organism physiology. We highlight the protein localization on the luminal surface, discussing important physiological receptors and the glycocalyx. The protein polarization to the abluminal membrane is especially unique in small resistance arteries with the presence of the myoendothelial junction, a signaling microdomain that regulates vasodilation, feedback to smooth muscle cells, and ultimately total peripheral resistance. We also discuss the interendothelial junction, where tight junctions, adherens junctions, and gap junctions all convene and regulate endothelial function. Finally, we address planar cell polarity, or axial polarity, and how this is regulated by mechanosensory signals like blood flow.
Collapse
Affiliation(s)
- Abigail G Wolpe
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA.,Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Claire A Ruddiman
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA.,Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Phillip J Hall
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA, .,Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, Virginia, USA,
| |
Collapse
|
19
|
Szulcek R, Grune J, Kuebler WM. The updated cancer paradigm of PAH: recognizing complexity. Am J Physiol Lung Cell Mol Physiol 2020; 318:L1111-L1114. [PMID: 32320264 DOI: 10.1152/ajplung.00159.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Affiliation(s)
- Robert Szulcek
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Heart Center Berlin, Berlin, Germany
| | - Jana Grune
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Centre for Cardiovascular Research (DZHK - partner site Berlin), Berlin, Germany
| | - Wolfgang M Kuebler
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Centre for Cardiovascular Research (DZHK - partner site Berlin), Berlin, Germany.,Departments of Surgery and Physiology, University of Toronto and Keenan Research Centre for Biomedical Science of St. Michael's, Toronto, Ontario, Canada
| |
Collapse
|
20
|
Obradovic M, Essack M, Zafirovic S, Sudar‐Milovanovic E, Bajic VP, Van Neste C, Trpkovic A, Stanimirovic J, Bajic VB, Isenovic ER. Redox control of vascular biology. Biofactors 2020; 46:246-262. [PMID: 31483915 PMCID: PMC7187163 DOI: 10.1002/biof.1559] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 08/14/2019] [Indexed: 12/12/2022]
Abstract
Redox control is lost when the antioxidant defense system cannot remove abnormally high concentrations of signaling molecules, such as reactive oxygen species (ROS). Chronically elevated levels of ROS cause oxidative stress that may eventually lead to cancer and cardiovascular and neurodegenerative diseases. In this review, we focus on redox effects in the vascular system. We pay close attention to the subcompartments of the vascular system (endothelium, smooth muscle cell layer) and give an overview of how redox changes influence those different compartments. We also review the core aspects of redox biology, cardiovascular physiology, and pathophysiology. Moreover, the topic-specific knowledgebase DES-RedoxVasc was used to develop two case studies, one focused on endothelial cells and the other on the vascular smooth muscle cells, as a starting point to possibly extend our knowledge of redox control in vascular biology.
Collapse
Affiliation(s)
- Milan Obradovic
- Laboratory of Radiobiology and Molecular GeneticsVinca Institute of Nuclear Sciences, University of BelgradeBelgradeSerbia
| | - Magbubah Essack
- King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Center (CBRC), Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE)ThuwalKingdom of Saudi Arabia
| | - Sonja Zafirovic
- Laboratory of Radiobiology and Molecular GeneticsVinca Institute of Nuclear Sciences, University of BelgradeBelgradeSerbia
| | - Emina Sudar‐Milovanovic
- Laboratory of Radiobiology and Molecular GeneticsVinca Institute of Nuclear Sciences, University of BelgradeBelgradeSerbia
| | - Vladan P. Bajic
- Laboratory of Radiobiology and Molecular GeneticsVinca Institute of Nuclear Sciences, University of BelgradeBelgradeSerbia
| | - Christophe Van Neste
- King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Center (CBRC), Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE)ThuwalKingdom of Saudi Arabia
| | - Andreja Trpkovic
- Laboratory of Radiobiology and Molecular GeneticsVinca Institute of Nuclear Sciences, University of BelgradeBelgradeSerbia
| | - Julijana Stanimirovic
- Laboratory of Radiobiology and Molecular GeneticsVinca Institute of Nuclear Sciences, University of BelgradeBelgradeSerbia
| | - Vladimir B. Bajic
- King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Center (CBRC), Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE)ThuwalKingdom of Saudi Arabia
| | - Esma R. Isenovic
- Laboratory of Radiobiology and Molecular GeneticsVinca Institute of Nuclear Sciences, University of BelgradeBelgradeSerbia
| |
Collapse
|
21
|
Chang JC. Sepsis and septic shock: endothelial molecular pathogenesis associated with vascular microthrombotic disease. Thromb J 2019; 17:10. [PMID: 31160889 PMCID: PMC6542012 DOI: 10.1186/s12959-019-0198-4] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 04/26/2019] [Indexed: 12/15/2022] Open
Abstract
In addition to protective “immune response”, sepsis is characterized by destructive “endothelial response” of the host, leading to endotheliopathy and its molecular dysfunction. Complement activation generates membrane attack complex (MAC). MAC causes channel formation to the cell membrane of pathogen, leading to death of microorganisms. In the host, MAC also may induce channel formation to innocent bystander endothelial cells (ECs) and ECs cannot be protected. This provokes endotheliopathy, which activates two independent molecular pathways: inflammatory and microthrombotic. Activated inflammatory pathway promotes the release of inflammatory cytokines and triggers inflammation. Activated microthrombotic pathway mediates platelet activation and exocytosis of unusually large von Willebrand factor multimers (ULVWF) from ECs and initiates microthrombogenesis. Excessively released ULVWF become anchored to ECs as long elongated strings and recruit activated platelets to assemble platelet-ULVWF complexes and form “microthrombi”. These microthrombi strings trigger disseminated intravascular microthrombosis (DIT), which is the underlying pathology of endotheliopathy-associated vascular microthrombotic disease (EA-VMTD). Sepsis-induced endotheliopathy promotes inflammation and DIT. Inflammation produces inflammatory response and DIT orchestrates consumptive thrombocytopenia, microangiopathic hemolytic anemia, and multiorgan dysfunction syndrome (MODS). Systemic inflammatory response syndrome (SIRS) is a combined phenotype of inflammation and endotheliopathy-associated (EA)-VMTD. Successful therapeutic design for sepsis can be achieved by counteracting the pathologic microthrombogenesis.
Collapse
Affiliation(s)
- Jae C Chang
- Department of Medicine, University of California Irvine School of Medicine, Irvine, CA USA
| |
Collapse
|
22
|
Esposito M, Mondal N, Greco TM, Wei Y, Spadazzi C, Lin SC, Zheng H, Cheung C, Magnani JL, Lin SH, Cristea IM, Sackstein R, Kang Y. Bone vascular niche E-selectin induces mesenchymal-epithelial transition and Wnt activation in cancer cells to promote bone metastasis. Nat Cell Biol 2019; 21:627-639. [PMID: 30988423 PMCID: PMC6556210 DOI: 10.1038/s41556-019-0309-2] [Citation(s) in RCA: 170] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 03/07/2019] [Indexed: 01/06/2023]
Abstract
How disseminated tumor cells (DTCs) engage specific stromal components in distant organs for survival and outgrowth is a critical but poorly understood step of the metastatic cascade. Previous studies have demonstrated the importance of the epithelial-mesenchymal transition (EMT) in promoting the cancer stem cell properties needed for metastasis initiation, while the reverse process of mesenchymal-epithelial transition (MET) is required for metastatic outgrowth. Here we report that this paradoxical requirement for simultaneous induction of both MET and cancer stem cell traits in DTCs is provided by bone vascular niche E-selectin, whose direct binding to cancer cells promotes bone metastasis by inducing MET and activating Wnt signaling. E-selectin binding activity mediated by α1–3 Fucosyltransferases Fut3/Fut6 and Glg1 are instrumental to the formation of bone metastasis. These findings provide unique insights into the functional role of E-selectin as a component of the vascular niche critical for metastatic colonization in bone.
Collapse
Affiliation(s)
- Mark Esposito
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Nandini Mondal
- Department of Dermatology and Harvard Skin Disease Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Program of Excellence in Glycosciences, Harvard Medical School, Boston, MA, USA
| | - Todd M Greco
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Yong Wei
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Chiara Spadazzi
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori IRCCS, Meldola, Italy
| | - Song-Chang Lin
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hanqiu Zheng
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Corey Cheung
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | | | - Sue-Hwa Lin
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ileana M Cristea
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Robert Sackstein
- Department of Dermatology and Harvard Skin Disease Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Program of Excellence in Glycosciences, Harvard Medical School, Boston, MA, USA.,Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
23
|
Weidenfeld S, Kuebler WM. Shedding First Light on the Alveolar Epithelial Glycocalyx. Am J Respir Cell Mol Biol 2018; 59:283-284. [DOI: 10.1165/rcmb.2018-0108ed] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Affiliation(s)
- Sarah Weidenfeld
- Institute of PhysiologyCharité–Universitätsmedizin BerlinBerlin, Germany
| | - Wolfgang M. Kuebler
- Institute of PhysiologyCharité–Universitätsmedizin BerlinBerlin, Germany
- Keenan Research Centre for Biomedical ScienceSt. Michael’s HospitalToronto, Canada
- Department of Surgeryand
- Department of PhysiologyUniversity of TorontoToronto, Canada
| |
Collapse
|
24
|
Simmons S, Erfinanda L, Bartz C, Kuebler WM. Novel mechanisms regulating endothelial barrier function in the pulmonary microcirculation. J Physiol 2018; 597:997-1021. [PMID: 30015354 DOI: 10.1113/jp276245] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 05/25/2018] [Indexed: 12/11/2022] Open
Abstract
The pulmonary epithelial and vascular endothelial cell layers provide two sequential physical and immunological barriers that together form a semi-permeable interface and prevent alveolar and interstitial oedema formation. In this review, we focus specifically on the continuous endothelium of the pulmonary microvascular bed that warrants strict control of the exchange of gases, fluid, solutes and circulating cells between the plasma and the interstitial space. The present review provides an overview of emerging molecular mechanisms that permit constant transcellular exchange between the vascular and interstitial compartment, and cause, prevent or reverse lung endothelial barrier failure under experimental conditions, yet with a clinical perspective. Based on recent findings and at times seemingly conflicting results we discuss emerging paradigms of permeability regulation by altered ion transport as well as shifts in the homeostasis of sphingolipids, angiopoietins and prostaglandins.
Collapse
Affiliation(s)
- Szandor Simmons
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Lasti Erfinanda
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Christoph Bartz
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Wolfgang M Kuebler
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada.,Departments of Surgery and Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
25
|
Robilliard LD, Kho DT, Johnson RH, Anchan A, O'Carroll SJ, Graham ES. The Importance of Multifrequency Impedance Sensing of Endothelial Barrier Formation Using ECIS Technology for the Generation of a Strong and Durable Paracellular Barrier. BIOSENSORS-BASEL 2018; 8:bios8030064. [PMID: 29973526 PMCID: PMC6163417 DOI: 10.3390/bios8030064] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 06/20/2018] [Accepted: 06/28/2018] [Indexed: 12/20/2022]
Abstract
In this paper, we demonstrate the application of electrical cell-substrate impedance sensing (ECIS) technology for measuring differences in the formation of a strong and durable endothelial barrier model. In addition, we highlight the capacity of ECIS technology to model the parameters of the physical barrier associated with (I) the paracellular space (referred to as Rb) and (II) the basal adhesion of the endothelial cells (α, alpha). Physiologically, both parameters are very important for the correct formation of endothelial barriers. ECIS technology is the only commercially available technology that can measure and model these parameters independently of each other, which is important in the context of ascertaining whether a change in overall barrier resistance (R) occurs because of molecular changes in the paracellular junctional molecules or changes in the basal adhesion molecules. Finally, we show that the temporal changes observed in the paracellular Rb can be associated with changes in specific junctional proteins (CD144, ZO-1, and catenins), which have major roles in governing the overall strength of the junctional communication between neighbouring endothelial cells.
Collapse
Affiliation(s)
- Laverne D Robilliard
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand.
- Department of Molecular Medicine and Pathology, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand.
| | - Dan T Kho
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand.
- Department of Molecular Medicine and Pathology, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand.
| | - Rebecca H Johnson
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand.
- Department of Molecular Medicine and Pathology, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand.
| | - Akshata Anchan
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand.
- Department of Molecular Medicine and Pathology, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand.
| | - Simon J O'Carroll
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand.
- Department of Anatomy and Medical Imaging, University of Auckland, Auckland 1023, New Zealand.
| | - Euan Scott Graham
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand.
- Department of Molecular Medicine and Pathology, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand.
| |
Collapse
|
26
|
Gao P, He C, Zhang C, Li B, Guo Y, Zhao W, Xie Q, Zhang X. Preventive effect on endothelial surface layer damage of Fusu agent in LPS-induced acute lung injury in rats. Mol Cell Biochem 2018; 450:113-123. [PMID: 29948507 PMCID: PMC6328519 DOI: 10.1007/s11010-018-3378-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 06/11/2018] [Indexed: 01/11/2023]
Abstract
Acute lung injury (ALI) is one of major causes of morbidity and mortality in intensive care. In pathophysiological events of ALI, endothelial surface layer (ESL) injury can result in capillary leakage as the initial event. The “Fusu agent”, a traditional Chinese medicine, can inhibit inflammatory factors, attenuate lung capillary leak as seen in our previous study. This study was aimed to explore the molecular mechanism of Fusu agent treatment with ALI. Consistent with previous studies, we found that Fusu agent has the protective effect on LPS-induced ALI model rats. Further investigation demonstrated that heparanase activation is necessary for the LPS-induced ALI model to aggravate ESL loss. Fusu agent can inhibit heparanase activation and heparan sulfate proteoglycans’ (HSPGs) degradation to mitigate the ESL injury. Furthermore, TNF-α and intercellular adhesion molecule-1 (ICAM-1) were significantly reduced upon Fusu agent pre-treatment to inhibit inflammatory cell influx and neutrophil adhesion in ALI. These findings shed light on the pharmacologic basis for the clinical application of traditional Chinese medicine in treating ALI.
Collapse
Affiliation(s)
- Peiyang Gao
- Critical Care Medicine, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Chengshi He
- Department of Respiratory, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chuantao Zhang
- Infectious Disease Department, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Baixue Li
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yiling Guo
- Department of Emergency, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wen Zhao
- Department of Emergency, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Quan Xie
- Critical Care Medicine, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xuemei Zhang
- Critical Care Medicine, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
27
|
Hilfenhaus G, Nguyen DP, Freshman J, Prajapati D, Ma F, Song D, Ziyad S, Cuadrado M, Pellegrini M, Bustelo XR, Iruela-Arispe ML. Vav3-induced cytoskeletal dynamics contribute to heterotypic properties of endothelial barriers. J Cell Biol 2018; 217:2813-2830. [PMID: 29858212 PMCID: PMC6080943 DOI: 10.1083/jcb.201706041] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 04/12/2018] [Accepted: 05/08/2018] [Indexed: 12/26/2022] Open
Abstract
Through multiple cell-cell and cell-matrix interactions, epithelial and endothelial sheets form tight barriers. Modulators of the cytoskeleton contribute to barrier stability and act as rheostats of vascular permeability. In this study, we sought to identify cytoskeletal regulators that underlie barrier diversity across vessels. To achieve this, we correlated functional and structural barrier features to gene expression of endothelial cells (ECs) derived from different vascular beds. Within a subset of identified candidates, we found that the guanosine nucleotide exchange factor Vav3 was exclusively expressed by microvascular ECs and was closely associated with a high-resistance barrier phenotype. Ectopic expression of Vav3 in large artery and brain ECs significantly enhanced barrier resistance and cortical rearrangement of the actin cytoskeleton. Mechanistically, we found that the barrier effect of Vav3 is dependent on its Dbl homology domain and downstream activation of Rap1. Importantly, inactivation of Vav3 in vivo resulted in increased vascular leakage, highlighting its function as a key regulator of barrier stability.
Collapse
Affiliation(s)
- Georg Hilfenhaus
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA
| | - Dai Phuong Nguyen
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA
| | - Jonathan Freshman
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA
| | - Divya Prajapati
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA
| | - Feiyang Ma
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA
| | - Dana Song
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA
| | - Safiyyah Ziyad
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA
| | - Myriam Cuadrado
- Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, and Centro de Investigación Biomédica en Red de Cáncer, Consejo Superior de Investigaciones Científicas, and University of Salamanca, Campus Unamuno, Salamanca, Spain
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA.,Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA
| | - Xosé R Bustelo
- Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, and Centro de Investigación Biomédica en Red de Cáncer, Consejo Superior de Investigaciones Científicas, and University of Salamanca, Campus Unamuno, Salamanca, Spain
| | - M Luisa Iruela-Arispe
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA .,Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA
| |
Collapse
|
28
|
A generic whole body physiologically based pharmacokinetic model for therapeutic proteins in PK-Sim. J Pharmacokinet Pharmacodyn 2017; 45:235-257. [PMID: 29234936 PMCID: PMC5845054 DOI: 10.1007/s10928-017-9559-4] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 12/05/2017] [Indexed: 12/24/2022]
Abstract
Proteins are an increasingly important class of drugs used as therapeutic as well as diagnostic agents. A generic physiologically based pharmacokinetic (PBPK) model was developed in order to represent at whole body level the fundamental mechanisms driving the distribution and clearance of large molecules like therapeutic proteins. The model was built as an extension of the PK-Sim model for small molecules incorporating (i) the two-pore formalism for drug extravasation from blood plasma to interstitial space, (ii) lymph flow, (iii) endosomal clearance and (iv) protection from endosomal clearance by neonatal Fc receptor (FcRn) mediated recycling as especially relevant for antibodies. For model development and evaluation, PK data was used for compounds with a wide range of solute radii. The model supports the integration of knowledge gained during all development phases of therapeutic proteins, enables translation from pre-clinical species to human and allows predictions of tissue concentration profiles which are of relevance for the analysis of on-target pharmacodynamic effects as well as off-target toxicity. The current implementation of the model replaces the generic protein PBPK model available in PK-Sim since version 4.2 and becomes part of the Open Systems Pharmacology Suite.
Collapse
|
29
|
Rayner SG, Zheng Y. Engineered Microvessels for the Study of Human Disease. J Biomech Eng 2017; 138:2545529. [PMID: 27537085 DOI: 10.1115/1.4034428] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Indexed: 12/22/2022]
Abstract
The microvasculature is an extensive, heterogeneous, and complex system that plays a critical role in human physiology and disease. It nourishes almost all living human cells and maintains a local microenvironment that is vital for tissue and organ function. Operating under a state of continuous flow, with an intricate architecture despite its small caliber, and subject to a multitude of biophysical and biochemical stimuli, the microvasculature can be a complex subject to study in the laboratory setting. Engineered microvessels provide an ideal platform that recapitulates essential elements of in vivo physiology and allows study of the microvasculature in a precise and reproducible way. Here, we review relevant structural and functional vascular biology, discuss different methods to engineer microvessels, and explore the applications of this exciting tool for the study of human disease.
Collapse
Affiliation(s)
- Samuel G Rayner
- Department of Pulmonary and Critical Care Medicine, University of Washington School of Medicine, Campus Box 356522, Seattle, WA 98195 e-mail:
| | - Ying Zheng
- Department of Bioengineering, University of Washington, 3720 15th Avenue NE, Seattle, WA 98105;Center for Cardiovascular Biology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109 e-mail:
| |
Collapse
|
30
|
Patra C, Boccaccini A, Engel F. Vascularisation for cardiac tissue engineering: the extracellular matrix. Thromb Haemost 2017; 113:532-47. [DOI: 10.1160/th14-05-0480] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Accepted: 09/03/2014] [Indexed: 02/07/2023]
Abstract
SummaryCardiovascular diseases present a major socio-economic burden. One major problem underlying most cardiovascular and congenital heart diseases is the irreversible loss of contractile heart muscle cells, the cardiomyocytes. To reverse damage incurred by myocardial infarction or by surgical correction of cardiac malformations, the loss of cardiac tissue with a thickness of a few millimetres needs to be compensated. A promising approach to this issue is cardiac tissue engineering. In this review we focus on the problem of in vitro vascularisation as implantation of cardiac patches consisting of more than three layers of cardiomyocytes (> 100 μm thick) already results in necrosis. We explain the need for vascularisation and elaborate on the importance to include non-myocytes in order to generate functional vascularised cardiac tissue. We discuss the potential of extracellular matrix molecules in promoting vascularisation and introduce nephronectin as an example of a new promising candidate. Finally, we discuss current biomaterial- based approaches including micropatterning, electrospinning, 3D micro-manufacturing technology and porogens. Collectively, the current literature supports the notion that cardiac tissue engineering is a realistic option for future treatment of paediatric and adult patients with cardiac disease.
Collapse
|
31
|
Khaddaj Mallat R, Mathew John C, Kendrick DJ, Braun AP. The vascular endothelium: A regulator of arterial tone and interface for the immune system. Crit Rev Clin Lab Sci 2017; 54:458-470. [PMID: 29084470 DOI: 10.1080/10408363.2017.1394267] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
As the primary interface between the blood and various tissues of the body, the vascular endothelium exhibits a diverse range of roles and activities, all of which contribute to the overall health and function of the cardiovascular system. In this focused review, we discuss several key aspects of endothelial function, how this may be compromised and subsequent consequences. Specifically, we examine the dynamic regulation of arterial contractility and distribution of blood flow through the generation of chemical and electrical signaling events that impinge upon vascular smooth muscle. The endothelium can generate a diverse range of vasoactive compounds and signals, most of which act locally to adjust blood flow in a dynamic fashion to match tissue metabolism. Disruption of these vascular signaling processes (e.g. reduced nitric oxide bioavailability) is typically referred to as endothelial dysfunction, which is a recognized risk factor for cardiovascular disease in patients and occurs early in the development and progression of hypertension, atherosclerosis and tissue ischemia. Endothelial dysfunction is also associated with type-2 Diabetes and aging and increased mechanistic knowledge of the cellular changes contributing to these effects may provide important clues for interventional strategies. The endothelium also serves as the initial site of interaction for immune cells entering tissues in response to damage and acts to facilitate the actions of both the innate and acquired immune systems to interact with the vascular wall. In addition to representing the main cell type responsible for the formation of new blood vessels (i.e. angiogenesis) within the vasculature, the endothelium is also emerging as a source of extracellular vesicle or microparticles for the transport of signaling molecules and other cellular materials to nearby, or remote, sites in the body. The characteristics of released microparticles appear to change with the functional status of the endothelium; thus, these microparticles may represent novel biomarkers of endothelial health and more serious cardiovascular disease.
Collapse
Affiliation(s)
- Rayan Khaddaj Mallat
- a Department of Physiology and Pharmacology, Cumming School of Medicine , University of Calgary, and Libin Cardiovascular Institute of Alberta , Calgary , Canada
| | - Cini Mathew John
- a Department of Physiology and Pharmacology, Cumming School of Medicine , University of Calgary, and Libin Cardiovascular Institute of Alberta , Calgary , Canada
| | - Dylan J Kendrick
- a Department of Physiology and Pharmacology, Cumming School of Medicine , University of Calgary, and Libin Cardiovascular Institute of Alberta , Calgary , Canada
| | - Andrew P Braun
- a Department of Physiology and Pharmacology, Cumming School of Medicine , University of Calgary, and Libin Cardiovascular Institute of Alberta , Calgary , Canada
| |
Collapse
|
32
|
Improved vessel painting with carbocyanine dye-liposome solution for visualisation of vasculature. Sci Rep 2017; 7:10089. [PMID: 28855543 PMCID: PMC5577039 DOI: 10.1038/s41598-017-09496-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 07/27/2017] [Indexed: 11/09/2022] Open
Abstract
Vessel painting is one of the most accessible and cost-effective techniques for visualising vasculature by fluorescence microscopy. In this method, the hydrophobic carbocyanine dye DiIC18 labels the plasma membrane via insertion of its alkyl chains into the lipid bilayer. A major disadvantage of this procedure is that it does not stain veins and some microvessels in mouse brain. Furthermore, DiIC18 molecules can aggregate during perfusion, thereby occluding arteries and reducing the success rate and reproducibility of the experiment. To overcome these problems, we developed an improved vessel painting procedure that employs neutral liposomes (NLs) and DiIC12. NLs prevented DiI aggregation under physiological conditions whereas DiIC12 showed enhanced dye incorporation into liposomes and consequently increased staining intensity. Using this method, we successfully labelled all major blood vessel types in the mouse brain, including both veins and microvessels. Thus, liposome-mediated vessel painting is a simple and efficient method for visualising vasculature.
Collapse
|
33
|
Piety NZ, Reinhart WH, Stutz J, Shevkoplyas SS. Optimal hematocrit in an artificial microvascular network. Transfusion 2017; 57:2257-2266. [PMID: 28681482 DOI: 10.1111/trf.14213] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 05/03/2017] [Accepted: 05/11/2017] [Indexed: 12/18/2022]
Abstract
BACKGROUND Higher hematocrit increases the oxygen-carrying capacity of blood but also increases blood viscosity, thus decreasing blood flow through the microvasculature and reducing the oxygen delivery to tissues. Therefore, an optimal value of hematocrit that maximizes tissue oxygenation must exist. STUDY DESIGN AND METHODS We used viscometry and an artificial microvascular network device to determine the optimal hematocrit in vitro. Suspensions of fresh red blood cells (RBCs) in plasma, normal saline, or a protein-containing buffer and suspensions of stored red blood cells (at Week 6 of standard hypothermic storage) in plasma with hematocrits ranging from 10 to 80% were evaluated. RESULTS For viscometry, optimal hematocrits were 10, 25.2, 31.9, 37.1, and 37.5% for fresh RBCs in plasma at shear rates of 3.2 or less, 11.0, 27.7, 69.5, and 128.5 inverse seconds. For the artificial microvascular network, optimal hematocrits were 51.1, 55.6, 59.2, 60.9, 62.3, and 64.6% for fresh RBCs in plasma and 46.4, 48.1, 54.8, 61.4, 65.7, and 66.5% for stored RBCs in plasma at pressures of 2.5, 5, 10, 20, 40, and 60 cm H2 O. CONCLUSION Although exact optimal hematocrit values may depend on specific microvascular architecture, our results suggest that the optimal hematocrit for oxygen delivery in the microvasculature depends on perfusion pressure. Therefore, anemia in chronic disorders may represent a beneficial physiological response to reduced perfusion pressure resulting from decreased heart function and/or vascular stenosis. Our results may help explain why a therapeutically increasing hematocrit in such conditions with RBC transfusion frequently leads to worse clinical outcomes.
Collapse
Affiliation(s)
- Nathaniel Z Piety
- Department of Biomedical Engineering, Cullen College of Engineering, University of Houston, Houston, Texas
| | | | - Julianne Stutz
- Department of Biomedical Engineering, Cullen College of Engineering, University of Houston, Houston, Texas
| | - Sergey S Shevkoplyas
- Department of Biomedical Engineering, Cullen College of Engineering, University of Houston, Houston, Texas
| |
Collapse
|
34
|
Peters EB. Endothelial Progenitor Cells for the Vascularization of Engineered Tissues. TISSUE ENGINEERING PART B-REVIEWS 2017; 24:1-24. [PMID: 28548628 DOI: 10.1089/ten.teb.2017.0127] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Self-assembled microvasculature from cocultures of endothelial cells (ECs) and stromal cells has significantly advanced efforts to vascularize engineered tissues by enhancing perfusion rates in vivo and producing investigative platforms for microvascular morphogenesis in vitro. However, to clinically translate prevascularized constructs, the issue of EC source must be resolved. Endothelial progenitor cells (EPCs) can be noninvasively supplied from the recipient through adult peripheral and umbilical cord blood, as well as derived from induced pluripotent stem cells, alleviating antigenicity issues. EPCs can also differentiate into all tissue endothelium, and have demonstrated potential for therapeutic vascularization. Yet, EPCs are not the standard EC choice to vascularize tissue constructs in vitro. Possible reasons include unresolved issues with EPC identity and characterization, as well as uncertainty in the selection of coculture, scaffold, and culture media combinations that promote EPC microvessel formation. This review addresses these issues through a summary of EPC vascular biology and the effects of tissue engineering design parameters upon EPC microvessel formation. Also included are perspectives to integrate EPCs with emerging technologies to produce functional, organotypic vascularized tissues.
Collapse
Affiliation(s)
- Erica B Peters
- Department of Chemical and Biological Engineering, University of Colorado , Boulder, Colorado
| |
Collapse
|
35
|
Burkhart A, Andresen TL, Aigner A, Thomsen LB, Moos T. Transfection of primary brain capillary endothelial cells for protein synthesis and secretion of recombinant erythropoietin: a strategy to enable protein delivery to the brain. Cell Mol Life Sci 2017; 74:2467-2485. [PMID: 28293718 PMCID: PMC11107693 DOI: 10.1007/s00018-017-2501-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 02/13/2017] [Accepted: 03/03/2017] [Indexed: 12/13/2022]
Abstract
Treatment of chronic disorders affecting the central nervous system (CNS) is complicated by the inability of drugs to cross the blood-brain barrier (BBB). Non-viral gene therapy applied to brain capillary endothelial cells (BCECs) denotes a novel approach to overcome the restraints in this passage, as turning BCECs into recombinant protein factories by transfection could result in protein secretion further into the brain. The present study aims to investigate the possibility of transfecting primary rat brain endothelial cells (RBECs) for recombinant protein synthesis and secretion of the neuroprotective protein erythropoietin (EPO). We previously showed that 4% of RBECs with BBB properties can be transfected without disrupting the BBB integrity in vitro, but it can be questioned whether this is sufficient to enable protein secretion at therapeutic levels. The present study examined various transfection vectors, with regard to increasing the transfection efficiency without disrupting the BBB integrity. Lipofectamine 3000™ was the most potent vector compared to polyethylenimine (PEI) and Turbofect. When co-cultured with astrocytes, the genetically modified RBECs secreted recombinant EPO into the cell culture medium both luminally and abluminally, and despite lower levels of EPO reaching the abluminal chamber, the amount of recombinant EPO was sufficient to evolve a biological effect on astrocytes cultured at the abluminal side in terms of upregulated gene expression of brain-derived neurotropic factor (BDNF). In conclusion, non-viral gene therapy to RBECs leads to protein secretion and signifies a method for therapeutic proteins to target cells inside the CNS otherwise omitted due to the BBB.
Collapse
Affiliation(s)
- Annette Burkhart
- Laboratory of Neurobiology, Biomedicine Group, Department of Health Science and Technology, Aalborg University, Frederik Bajers Vej 3B, 2.104, 9220, Aalborg East, Denmark.
| | - Thomas Lars Andresen
- DTU Nanotech, Technical University of Denmark, Produktionstorvet Building 423, 2800, Kongens Lyngby, Denmark
| | - Achim Aigner
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, University of Leipzig, Härtelstraße 16-18, 04107, Leipzig, Germany
| | - Louiza Bohn Thomsen
- Laboratory of Neurobiology, Biomedicine Group, Department of Health Science and Technology, Aalborg University, Frederik Bajers Vej 3B, 2.104, 9220, Aalborg East, Denmark
| | - Torben Moos
- Laboratory of Neurobiology, Biomedicine Group, Department of Health Science and Technology, Aalborg University, Frederik Bajers Vej 3B, 2.104, 9220, Aalborg East, Denmark
| |
Collapse
|
36
|
Iba T, Sasaki T, Ohshima K, Sato K, Nagaoka I, Thachil J. The Comparison of the Protective Effects of α- and β-Antithrombin against Vascular Endothelial Cell Damage Induced by Histone in Vitro. TH OPEN 2017; 1:e3-e10. [PMID: 31249909 PMCID: PMC6524836 DOI: 10.1055/s-0037-1603926] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Antithrombin is a promising option for the treatment of sepsis, and vascular endothelium is an important target for this fatal condition. Here, we aimed to evaluate the protective effects of different glycoforms of antithrombin on histone-induced endothelial cell damage and explore the responsible mechanisms in an experimental model in vitro. Endothelial cells were treated in vitro using histone H4 to induce cellular damage. Various doses of either α- or β-antithrombin were used as treatment interventions, and both cell viability and the levels of lactate dehydrogenase (LDH) in the medium were assessed. Endothelial cell damage was also assessed using microscopic examination and immunofluorescent staining with anti-syndecan-4 and anti-antithrombin antibodies. As a result, both glycoforms of antithrombin significantly improved cell viability when administered at a physiological dose (150 μg/mL). Cellular injury as evaluated using the LDH level was significantly suppressed by β-antithrombin at a supranormal dose (600 μg/mL). Microscopic observation suggested that β-antithrombin suppressed the endothelial cell damage more efficiently than α-antithrombin. β-Antithrombin suppressed the intensity of syndecan-4 staining which became evident after treatment with histone H4, more prominently than α-antithrombin. The distribution of antithrombin was identical to that of syndecan-4. In conclusion, both α- and β-antithrombin can protect vascular endothelial cells from histone H4-induced damage, although the effect was stronger for β-antithrombin. The responsible mechanisms might involve the binding of antithrombin to the glycocalyx on the endothelial surface. These results provide a theoretical basis for the application of antithrombin to the prevention and treatment of sepsis-related endothelial damage.
Collapse
Affiliation(s)
- Toshiaki Iba
- Department of Emergency and Disaster Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Tetsuya Sasaki
- Nihon Pharmaceutical Co. Ltd., Research Laboratory, Narita, Japan
| | | | - Koichi Sato
- Department of Surgery, Juntendo Shizuoka Hospital, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Isao Nagaoka
- Department of Host Defense and Biochemical Research, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Jecko Thachil
- Department of Haematology, Manchester Royal Infirmary, Manchester, United Kingdom
| |
Collapse
|
37
|
Daiber A, Steven S, Weber A, Shuvaev VV, Muzykantov VR, Laher I, Li H, Lamas S, Münzel T. Targeting vascular (endothelial) dysfunction. Br J Pharmacol 2017; 174:1591-1619. [PMID: 27187006 PMCID: PMC5446575 DOI: 10.1111/bph.13517] [Citation(s) in RCA: 335] [Impact Index Per Article: 41.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 04/28/2016] [Accepted: 05/09/2016] [Indexed: 12/18/2022] Open
Abstract
Cardiovascular diseases are major contributors to global deaths and disability-adjusted life years, with hypertension a significant risk factor for all causes of death. The endothelium that lines the inner wall of the vasculature regulates essential haemostatic functions, such as vascular tone, circulation of blood cells, inflammation and platelet activity. Endothelial dysfunction is an early predictor of atherosclerosis and future cardiovascular events. We review the prognostic value of obtaining measurements of endothelial function, the clinical techniques for its determination, the mechanisms leading to endothelial dysfunction and the therapeutic treatment of endothelial dysfunction. Since vascular oxidative stress and inflammation are major determinants of endothelial function, we have also addressed current antioxidant and anti-inflammatory therapies. In the light of recent data that dispute the prognostic value of endothelial function in healthy human cohorts, we also discuss alternative diagnostic parameters such as vascular stiffness index and intima/media thickness ratio. We also suggest that assessing vascular function, including that of smooth muscle and even perivascular adipose tissue, may be an appropriate parameter for clinical investigations. LINKED ARTICLES This article is part of a themed section on Redox Biology and Oxidative Stress in Health and Disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.12/issuetoc.
Collapse
Affiliation(s)
- Andreas Daiber
- Center of CardiologyMedical Center of the Johannes Gutenberg UniversityMainzGermany
- German Center for Cardiovascular Research (DZHK)Partner Site Rhine‐MainMainzGermany
| | - Sebastian Steven
- Center of CardiologyMedical Center of the Johannes Gutenberg UniversityMainzGermany
- Center of Thrombosis and HemostasisMedical Center of the Johannes Gutenberg UniversityMainzGermany
| | - Alina Weber
- Center of CardiologyMedical Center of the Johannes Gutenberg UniversityMainzGermany
| | - Vladimir V. Shuvaev
- Department of Systems Pharmacology & Translational Therapeutics, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Vladimir R. Muzykantov
- Department of Systems Pharmacology & Translational Therapeutics, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Ismail Laher
- Department of Pharmacology and Therapeutics, Faculty of MedicineUniversity of British ColumbiaVancouverBCCanada
| | - Huige Li
- German Center for Cardiovascular Research (DZHK)Partner Site Rhine‐MainMainzGermany
- Department of PharmacologyMedical Center of the Johannes Gutenberg UniversityMainzGermany
| | - Santiago Lamas
- Department of Cell Biology and ImmunologyCentro de Biología Molecular "Severo Ochoa" (CSIC‐UAM)MadridSpain
| | - Thomas Münzel
- Center of CardiologyMedical Center of the Johannes Gutenberg UniversityMainzGermany
- German Center for Cardiovascular Research (DZHK)Partner Site Rhine‐MainMainzGermany
| |
Collapse
|
38
|
Kuebler WM. The Flow-Dependent Transcription Factor KLF2 Protects Lung Vascular Barrier Function in Acute Respiratory Distress Syndrome. Am J Respir Crit Care Med 2017; 195:553-555. [DOI: 10.1164/rccm.201609-1946ed] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Wolfgang M. Kuebler
- Institute of PhysiologyCharité–Universitaetsmedizin BerlinBerlin, Germany
- The Keenan Research Centre for Biomedical Science at St. Michael’sToronto, Ontario, Canada
- Department of Surgeryand
- Department of PhysiologyUniversity of TorontoToronto, Ontario, Canada
| |
Collapse
|
39
|
Dumnicka P, Maduzia D, Ceranowicz P, Olszanecki R, Drożdż R, Kuśnierz-Cabala B. The Interplay between Inflammation, Coagulation and Endothelial Injury in the Early Phase of Acute Pancreatitis: Clinical Implications. Int J Mol Sci 2017; 18:E354. [PMID: 28208708 PMCID: PMC5343889 DOI: 10.3390/ijms18020354] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 01/17/2017] [Accepted: 01/31/2017] [Indexed: 02/07/2023] Open
Abstract
Acute pancreatitis (AP) is an inflammatory disease with varied severity, ranging from mild local inflammation to severe systemic involvement resulting in substantial mortality. Early pathologic events in AP, both local and systemic, are associated with vascular derangements, including endothelial activation and injury, dysregulation of vasomotor tone, increased vascular permeability, increased leukocyte migration to tissues, and activation of coagulation. The purpose of the review was to summarize current evidence regarding the interplay between inflammation, coagulation and endothelial dysfunction in the early phase of AP. Practical aspects were emphasized: (1) we summarized available data on diagnostic usefulness of the markers of endothelial dysfunction and activated coagulation in early prediction of severe AP; (2) we reviewed in detail the results of experimental studies and clinical trials targeting coagulation-inflammation interactions in severe AP. Among laboratory tests, d-dimer and angiopoietin-2 measurements seem the most useful in early prediction of severe AP. Although most clinical trials evaluating anticoagulants in treatment of severe AP did not show benefits, they also did not show significantly increased bleeding risk. Promising results of human trials were published for low molecular weight heparin treatment. Several anticoagulants that proved beneficial in animal experiments are thus worth testing in patients.
Collapse
Affiliation(s)
- Paulina Dumnicka
- Department of Medical Diagnostics, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland.
| | - Dawid Maduzia
- Department of Anatomy, Jagiellonian University Medical College, Kopernika 12, 31-034 Kraków, Poland.
| | - Piotr Ceranowicz
- Department of Physiology, Jagiellonian University Medical College, Grzegórzecka 16, 31-531 Kraków, Poland.
| | - Rafał Olszanecki
- Department of Pharmacology, Jagiellonian University Medical College, Grzegórzecka 16, 31-531 Kraków, Poland.
| | - Ryszard Drożdż
- Department of Medical Diagnostics, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland.
| | - Beata Kuśnierz-Cabala
- Department of Diagnostics, Chair of Clinical Biochemistry, Jagiellonian University Medical College, Kopernika 15A, 31-501 Kraków, Poland.
| |
Collapse
|
40
|
Belvitch P, Brown ME, Brinley BN, Letsiou E, Rizzo AN, Garcia JGN, Dudek SM. The ARP 2/3 complex mediates endothelial barrier function and recovery. Pulm Circ 2017; 7:200-210. [PMID: 28680579 PMCID: PMC5448540 DOI: 10.1086/690307] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 11/23/2016] [Indexed: 01/03/2023] Open
Abstract
Pulmonary endothelial cell (EC) barrier dysfunction and recovery is critical to the pathophysiology of acute respiratory distress syndrome. Cytoskeletal and subsequent cell membrane dynamics play a key mechanistic role in determination of EC barrier integrity. Here, we characterizAQe the actin related protein 2/3 (Arp 2/3) complex, a regulator of peripheral branched actin polymerization, in human pulmonary EC barrier function through studies of transendothelial electrical resistance (TER), intercellular gap formation, peripheral cytoskeletal structures and lamellipodia. Compared to control, Arp 2/3 inhibition with the small molecule inhibitor CK-666 results in a reduction of baseline barrier function (1,241 ± 53 vs 988 ± 64 ohm; p < 0.01), S1P-induced barrier enhancement and delayed recovery of barrier function after thrombin (143 ± 14 vs 93 ± 6 min; p < 0.01). Functional changes of Arp 2/3 inhibition on barrier integrity are associated temporally with increased intercellular gap area at baseline (0.456 ± 0.02 vs 0.299 ± 0.02; p < 0.05) and thirty minutes after thrombin (0.885 ± 0.03 vs 0.754 ± 0.03; p < 0.05). Immunofluorescent microscopy reveals reduced lamellipodia formation after S1P and during thrombin recovery in Arp 2/3 inhibited cells. Individual lamellipodia demonstrate reduced depth following Arp 2/3 inhibition vs vehicle at baseline (1.83 ± 0.41 vs 2.55 ± 0.46 µm; p < 0.05) and thirty minutes after S1P treatment (1.53 ± 0.37 vs 2.09 ± 0.36 µm; p < 0.05). These results establish a critical role for Arp 2/3 activity in determination of pulmonary endothelial barrier function and recovery through formation of EC lamellipodia and closure of intercellular gaps.
Collapse
Affiliation(s)
- Patrick Belvitch
- Division of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois Hospital and Health Science System, Chicago, IL, USA
| | - Mary E Brown
- Division of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois Hospital and Health Science System, Chicago, IL, USA
| | | | - Eleftheria Letsiou
- Division of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois Hospital and Health Science System, Chicago, IL, USA
| | - Alicia N Rizzo
- Division of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois Hospital and Health Science System, Chicago, IL, USA
| | - Joe G N Garcia
- University of Arizona Health Sciences Center, Tucson, AZ, USA
| | - Steven M Dudek
- Division of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois Hospital and Health Science System, Chicago, IL, USA
| |
Collapse
|
41
|
Abstract
Over 50% of the human body is comprised of fluids that are distributed in defined compartments. Although compartmentalized, these fluids are dynamically connected. Fluids, electrolytes, and acid-base balance in each compartment are tightly regulated, mostly in an energy-dependent manner to achieve their designed functions. For over a century, our understanding of the microvascular fluid homeostasis has evolved from hypothesized Ernest Starling principle to evidence-based and the revised Starling principle, incorporating the functional endothelial surface layer. The kidney is a highly vascular and encapsulated organ that is exquisitely sensitive to inadequate (insufficient or excess) blood flow. The kidney is particularly sensitive to venous congestion, and studies show that reduced venous return triggers a greater degree of kidney damage than that from lacking arterial flow. Thus, fluid overload can induce severe and sustained kidney injury. In the setting of established acute kidney injury, fluid management can be challenging. Impaired capacity of urine output and urine concentration and dilution should be taken into consideration when designing fluid therapy. Video Journal Club 'Cappuccino with Claudio Ronco' at http://www.karger.com/?doi=452702.
Collapse
Affiliation(s)
- Xiaoqiang Ding
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | | | | |
Collapse
|
42
|
Long DS, Hou W, Taylor RS, McCowan LME. Serum levels of endothelial glycocalyx constituents in women at 20 weeks' gestation who later develop gestational diabetes mellitus compared to matched controls: a pilot study. BMJ Open 2016; 6:e011244. [PMID: 27979833 PMCID: PMC5168613 DOI: 10.1136/bmjopen-2016-011244] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVES The aim of this pilot study was to determine the serum concentration of heparan sulfate, hyaluronan, chondroitin sulfate and syndecan-1 and if these serum concentrations can be used to identify women at 20 weeks' gestation who later develop gestational diabetes mellitus (GDM). DESIGN Nested case-control study from Auckland, New Zealand participants in the prospective cohort Screening for Pregnancy Endpoints study. SETTING Auckland, New Zealand. PARTICIPANTS 20 pregnant women (70% European, 15% Indian, 10% Asian, 5% Pacific Islander) at 20 weeks' gestation without any hypertensive complications who developed GDM by existing New Zealand criteria defined as a fasting glucose ≥5.5 mmol/L and/or 2 hours ≥9.0 mmol/L after a 75 g Oral Glucose Tolerance Test. Women not meeting these criteria were excluded from this study. The patients with GDM were matched with 20 women who had uncomplicated pregnancies and negative screening for GDM and matched for ethnicity, maternal age and BMI. PRIMARY AND SECONDARY OUTCOME MEASURES The primary measures were the serum concentrations of syndecan-1, heparan sulfate, hyaluronan and chondroitin sulfate determined by quantitative ELISA. There were no secondary outcome measures. RESULTS Binary logistic regression was performed to determine if serum concentrations of endothelial glycocalyx layer constituents in women at 20 weeks' gestation would be useful in predicting the subsequent diagnosis of GDM. The model was not statistically significant χ2=12.5, df=8, p=0.13, which indicates that the model was unable to distinguish between pregnant women at 20 weeks' gestation who later developed GDM and those who did not. CONCLUSIONS Serum concentrations of syndecan-1, heparan sulfate, hyaluronan and chondroitin sulfate in pregnant women at 20 weeks' gestation were not associated with later development of GDM. To further explore whether there is any relationship between endothelial glycocalyx constituents and GDM, the next step is to evaluate serum concentrations at the time diagnosis of GDM.
Collapse
Affiliation(s)
- David S Long
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
- Department of Engineering Science, University of Auckland, Auckland, New Zealand
| | - Weilin Hou
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Rennae S Taylor
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
| | - Lesley M E McCowan
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
| |
Collapse
|
43
|
Abstract
In the United States trauma is the leading cause of mortality among those under the age of 45, claiming approximately 192,000 lives each year. Significant personal disability, lost productivity, and long-term healthcare needs are common and contribute 580 billion dollars in economic impact each year. Improving resuscitation strategies and the early acute care of trauma patients has the potential to reduce the pathological sequelae of combined exuberant inflammation and immune suppression that can co-exist, or occur temporally, and adversely affect outcomes. The endothelial and epithelial glycocalyx has emerged as an important participant in both inflammation and immunomodulation. Constituents of the glycocalyx have been used as biomarkers of injury severity and have the potential to be target(s) for therapeutic interventions aimed at immune modulation. In this review, we provide a contemporary understanding of the physiologic structure and function of the glycocalyx and its role in traumatic injury with a particular emphasis on lung injury.
Collapse
|
44
|
Perioperative fluid therapy: defining a clinical algorithm between insufficient and excessive. J Clin Anesth 2016; 35:384-391. [DOI: 10.1016/j.jclinane.2016.08.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 08/09/2016] [Indexed: 01/03/2023]
|
45
|
|
46
|
Composition and Function of the Interstitial Fluid. Protein Sci 2016. [DOI: 10.1201/9781315374307-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
47
|
Čejková S, Králová Lesná I, Poledne R. Monocyte adhesion to the endothelium is an initial stage of atherosclerosis development. COR ET VASA 2016. [DOI: 10.1016/j.crvasa.2015.08.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
48
|
Endothélium et microcirculation au cours des états critiques. Actes du séminaire de recherche translationnelle de la Société de réanimation de langue française (1er décembre 2015). MEDECINE INTENSIVE REANIMATION 2016. [DOI: 10.1007/s13546-016-1190-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
49
|
Piperigkou Z, Karamanou K, Engin AB, Gialeli C, Docea AO, Vynios DH, Pavão MS, Golokhvast KS, Shtilman MI, Argiris A, Shishatskaya E, Tsatsakis AM. Emerging aspects of nanotoxicology in health and disease: From agriculture and food sector to cancer therapeutics. Food Chem Toxicol 2016; 91:42-57. [DOI: 10.1016/j.fct.2016.03.003] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 03/02/2016] [Accepted: 03/03/2016] [Indexed: 02/07/2023]
|
50
|
Francis M, Xu N, Zhou C, Stevens T. Transient Receptor Potential Channel 4 Encodes a Vascular Permeability Defect and High-Frequency Ca(2+) Transients in Severe Pulmonary Arterial Hypertension. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:1701-9. [PMID: 27083517 DOI: 10.1016/j.ajpath.2016.02.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 01/28/2016] [Accepted: 02/01/2016] [Indexed: 11/19/2022]
Abstract
The canonical transient receptor potential channel 4 (TRPC4) comprises an endothelial store-operated Ca(2+) entry channel, and TRPC4 inactivation confers a survival benefit in pulmonary arterial hypertension (PAH). Endothelial Ca(2+) signals mediated by TRPC4 enhance vascular permeability in vitro, but the contribution of TRPC4-dependent Ca(2+) signals to the regulation of endothelial permeability in PAH is poorly understood. We tested the hypothesis that TRPC4 increases vascular permeability and alters the frequency of endothelial Ca(2+) transients in PAH. We measured permeability in isolated lungs, and found that TRPC4 exaggerated permeability responses to thapsigargin in Sugen/hypoxia-treated PAH rats. We compared endothelial Ca(2+) activity of wild-type with TRPC4-knockout rats using confocal microscopy, and evaluated how Ca(2+) signals were influenced in response to thapsigargin and sequential treatment with acetylcholine. We found that thapsigargin-stimulated Ca(2+) signals were increased in PAH, and recovered by TRPC4 inactivation. Store depletion revealed bimodal Ca(2+) responses to acetylcholine, with both short- and long-duration populations. Our results show that TRPC4 underlies an exaggerated endothelial permeability response in PAH. Furthermore, TRPC4 increased the frequency of endothelial Ca(2+) transients in severe PAH, suggesting that TRPC4 provides a Ca(2+) source associated with endothelial dysfunction in the pathophysiology of PAH. This phenomenon represents a new facet of the etiology of PAH, and may contribute to PAH vasculopathy by enabling inflammatory mediator flux across the endothelial barrier.
Collapse
Affiliation(s)
- Michael Francis
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama; Center for Lung Biology, University of South Alabama, Mobile, Alabama
| | - Ningyong Xu
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama; Center for Lung Biology, University of South Alabama, Mobile, Alabama
| | - Chun Zhou
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama; Center for Lung Biology, University of South Alabama, Mobile, Alabama
| | - Troy Stevens
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama; Center for Lung Biology, University of South Alabama, Mobile, Alabama; Department of Medicine, University of South Alabama, Mobile, Alabama.
| |
Collapse
|