1
|
Liu D, Liao P, Li H, Tong S, Wang B, Lu Y, Gao Y, Huang Y, Zhou H, Shi L, Papadimitriou J, Zong Y, Yuan J, Chen P, Chen Z, Ding P, Zheng Y, Zhang C, Zheng M, Gao J. Regulation of blood-brain barrier integrity by Dmp1-expressing astrocytes through mitochondrial transfer. SCIENCE ADVANCES 2024; 10:eadk2913. [PMID: 38941455 PMCID: PMC11212732 DOI: 10.1126/sciadv.adk2913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 05/24/2024] [Indexed: 06/30/2024]
Abstract
The blood-brain barrier (BBB) acts as the crucial physical filtration structure in the central nervous system. Here, we investigate the role of a specific subset of astrocytes in the regulation of BBB integrity. We showed that Dmp1-expressing astrocytes transfer mitochondria to endothelial cells via their endfeet for maintaining BBB integrity. Deletion of the Mitofusin 2 (Mfn2) gene in Dmp1-expressing astrocytes inhibited the mitochondrial transfer and caused BBB leakage. In addition, the decrease of MFN2 in astrocytes contributes to the age-associated reduction of mitochondrial transfer efficiency and thus compromises the integrity of BBB. Together, we describe a mechanism in which astrocytes regulate BBB integrity through mitochondrial transfer. Our findings provide innnovative insights into the cellular framework that underpins the progressive breakdown of BBB associated with aging and disease.
Collapse
Affiliation(s)
- Delin Liu
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, Western Australia 6009, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, Western Australia 6009, Australia
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Peng Liao
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Hao Li
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Sihan Tong
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Bingqi Wang
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yafei Lu
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Youshui Gao
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yigang Huang
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Hao Zhou
- Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310009, China
| | - Linjing Shi
- Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310009, China
| | - John Papadimitriou
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, Western Australia 6009, Australia
- Department of Pathology, Pathwest, Nedlands, Western Australia 6009, Australia
| | - Yao Zong
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, Western Australia 6009, Australia
| | - Jun Yuan
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, Western Australia 6009, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, Western Australia 6009, Australia
| | - Peilin Chen
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, Western Australia 6009, Australia
| | - Ziming Chen
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, Western Australia 6009, Australia
| | - Peng Ding
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yongqiang Zheng
- Department of Orthopaedics, Jinjiang Municipal Hospital, Jinjiang, Fujian Province, 362200, China
| | - Changqing Zhang
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Minghao Zheng
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, Western Australia 6009, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, Western Australia 6009, Australia
| | - Junjie Gao
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Department of Orthopaedics, Jinjiang Municipal Hospital, Jinjiang, Fujian Province, 362200, China
| |
Collapse
|
2
|
Cherniavska Y, Davydenko A, Pokhylko V, Fishchuk L, Rossokha Z. Preliminary study of the influence of maternal and neonatal NOS3 (rs1799983), IL1B (rs1143634) genes variants and their intergenic interaction on the development of hypoxic-ischemic encephalopathy in newborns in the context of treatment planning. WIADOMOSCI LEKARSKIE (WARSAW, POLAND : 1960) 2024; 77:2373-2380. [PMID: 39874319 DOI: 10.36740/wlek/197108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2025]
Abstract
OBJECTIVE Aim: To determine the influence of maternal and neonatal variants of the eNOS (G894T, rs1799983) and IL1B (C3953T, rs1143634) genes and their intergenic interactions on the development of HIE in newborns. PATIENTS AND METHODS Materials and Methods: The study included a cohort of 105 newborns and their 99 mothers. Determination of variants of the genes eNOS (G894T, rs1799983) and IL1B (C3953T, rs1143634) was carried out for the patients of study groups. RESULTS Results: The frequency of detection of the 894TT genotype by the eNOS gene was increased in newborns with severe asphyxia (p=0.018) and in their mothers (p=0,0057). Further analysis of intergenic interactions, performed in mother-child pairs, revealed an increased frequency of the neonatal 894GG (eNOS)/maternal 3953С (IL-1B) genotype combination in the comparison group versus the group of newborns with HIE (p=0.007). CONCLUSION Conclusions: The significance of the intergenic maternal combination of 894GG/3953CT genotypes for the eNOS and IL1B genes and the intergenic combination of neonatal 894GG (eNOS)/maternal 3953CT (IL-1B) genotypes in the development of HIE in newborns has been proven. Associations of maternal and neonatal 894TT genotypes for the eNOS gene with the development of severe asphyxia, bradycardia, and respiratory failure were found in newborns with HIE.
Collapse
Affiliation(s)
- Yuliia Cherniavska
- DEPARTMENT OF PEDIATRICS NO1 AND NEONATOLOGY, POLTAVA STATE MEDICAL UNIVERSITY, POLTAVA, UKRAINE
| | - Alina Davydenko
- DEPARTMENT OF ANESTHESIOLOGY AND INTENSIVE CARE, POLTAVA STATE MEDICAL UNIVERSITY, POLTAVE, UKRAINE
| | - Valerii Pokhylko
- DEPARTMENT OF PEDIATRICS NO1 AND NEONATOLOGY, POLTAVA STATE MEDICAL UNIVERSITY, POLTAVA, UKRAINE
| | - Liliia Fishchuk
- EXPERT-ANALYTICAL MEDICAL CENTER FOR MOLECULAR GENETICS, SHUPYK NATIONAL HEALTHCARE UNIVERSITY OF UKRAINE, KYIV, UKRAINE
| | - Zoia Rossokha
- EXPERT-ANALYTICAL MEDICAL CENTER FOR MOLECULAR GENETICS, SHUPYK NATIONAL HEALTHCARE UNIVERSITY OF UKRAINE, KYIV, UKRAINE
| |
Collapse
|
3
|
Longitudinal Assessment of Plasma Syndecan-1 Predicts 60-Day Mortality in Patients with COVID-19. J Clin Med 2023; 12:jcm12020552. [PMID: 36675479 PMCID: PMC9865511 DOI: 10.3390/jcm12020552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 01/03/2023] [Accepted: 01/04/2023] [Indexed: 01/12/2023] Open
Abstract
Background: Endotheliopathy is a common pathologic finding in patients with acute and long COVID-19. It may be associated with disease severity and predispose patients to long-term complications. Plasma levels of a proteoglycan, syndecan-1, are found to be significantly elevated in patients with COVID-19, but its roles in assessing disease severity and predicting long-term outcome are not fully understood. Methods: A total of 124 consecutive hospitalized patients with SARS-CoV-2 infection were prospectively enrolled and blood samples were collected on admission (T1), 3−4 days following treatment (T2), and 1−2 days prior to discharge or death (T3). Plasma levels of syndecan-1 were determined using an immunosorbent assay; various statistical analyses were performed to determine the association between plasma syndecan-1 levels and disease severity or the 60-day mortality rate. Results: Compared with those in the healthy controls, plasma levels of syndecan-1 in patients with critical COVID-19 were significantly higher (p < 0.0001). However, there was no statistically significant difference among patients with different disease severity (p > 0.05), resulting from large individual variability. Longitudinal analysis demonstrated that while the levels fluctuated during hospitalization in all patients, plasma syndecan-1 levels were persistently elevated from baseline in critical COVID-19 patients. Cox proportional hazard regression analyses revealed that elevated plasma levels of syndecan-1 (>260 ng/mL at T1, >1018 ng/mL at T2, and >461 ng/mL at T3) were significantly associated with the 60-day mortality rate. Conclusions: Endotheliopathy, marked by glycocalyx degradation and elevated plasma syndecan-1, occurs in nearly all hospitalized patients with SARS-CoV-2 infection; elevated plasma syndecan-1 is associated with increased mortality in COVID-19 patients.
Collapse
|
4
|
H 2S protects hippocampal neurons against hypoxia-reoxygenation injury by promoting RhoA phosphorylation at Ser188. Cell Death Discov 2021; 7:132. [PMID: 34088899 PMCID: PMC8178328 DOI: 10.1038/s41420-021-00514-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/12/2021] [Accepted: 05/01/2021] [Indexed: 12/11/2022] Open
Abstract
Inhibition of RhoA-ROCK pathway is involved in the H2S-induced cerebral vasodilatation and H2S-mediated protection on endothelial cells against oxygen-glucose deprivation/reoxygenation injury. However, the inhibitory mechanism of H2S on RhoA-ROCK pathway is still unclear. The aim of this study was to investigate the target and mechanism of H2S in inhibition of RhoA/ROCK. GST-RhoAwild and GST-RhoAS188A proteins were constructed and expressed, and were used for phosphorylation assay in vitro. Recombinant RhoAwild-pEGFP-N1 and RhoAS188A-pEGFP-N1 plasmids were constructed and transfected into primary hippocampal nerve cells (HNCs) to evaluate the neuroprotective mechanism of endothelial H2S by using transwell co-culture system with endothelial cells from cystathionine-γ-lyase knockout (CSE-/-) mice and 3-mercaptopyruvate sulfurtransferase knockout (3-MST-/-) rats, respectively. We found that NaHS, exogenous H2S donor, promoted RhoA phosphorylation at Ser188 in the presence of cGMP-dependent protein kinase 1 (PKG1) in vitro. Besides, both exogenous and endothelial H2S facilitated the RhoA phosphorylation at Ser188 in HNCs, which induced the reduction of RhoA activity and membrane transposition, as well as ROCK2 activity and expression. To further investigate the role of endothelial H2S on RhoA phosphorylation, we detected H2S release from ECs of CSE+/+ and CSE-/- mice, and 3-MST+/+ and 3-MST-/- rats, respectively, and found that H2S produced by ECs in the culture medium is mainly catalyzed by CSE synthase. Moreover, we revealed that both endothelial H2S, mainly catalyzed by CSE, and exogenous H2S protected the HNCs against hypoxia-reoxygenation injury via phosphorylating RhoA at Ser188.
Collapse
|
5
|
Lynch CE, Eisenbaum M, Algamal M, Balbi M, Ferguson S, Mouzon B, Saltiel N, Ojo J, Diaz-Arrastia R, Mullan M, Crawford F, Bachmeier C. Impairment of cerebrovascular reactivity in response to hypercapnic challenge in a mouse model of repetitive mild traumatic brain injury. J Cereb Blood Flow Metab 2021; 41:1362-1378. [PMID: 33050825 PMCID: PMC8142124 DOI: 10.1177/0271678x20954015] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Incidences of repetitive mild TBI (r-mTBI), like those sustained by contact sports athletes and military personnel, are thought to be a risk factor for development of neurodegenerative disorders. Those suffering from chronic TBI-related illness demonstrate deficits in cerebrovascular reactivity (CVR), the ability of the cerebral vasculature to respond to a vasoactive stimulus. CVR is thus an important measure of traumatic cerebral vascular injury (TCVI), and a possible in vivo endophenotype of TBI-related neuropathogenesis. We combined laser speckle imaging of CVR in response to hypercapnic challenge with neurobehavioral assessment of learning and memory, to investigate if decreased cerebrovascular responsiveness underlies impaired cognitive function in our mouse model of chronic r-mTBI. We demonstrate a profile of blunted hypercapnia-evoked CVR in the cortices of r-mTBI mice like that of human TBI, alongside sustained memory and learning impairment, without biochemical or immunohistopathological signs of cerebral vessel laminar or endothelium constituent loss. Transient decreased expression of alpha smooth muscle actin and platelet-derived growth factor receptor β, indicative of TCVI, is obvious only at the time of the most pronounced CVR deficit. These findings implicate CVR as a valid preclinical measure of TCVI, perhaps useful for developing therapies targeting TCVI after recurrent mild head trauma.
Collapse
Affiliation(s)
- Cillian E Lynch
- The Roskamp Institute, Sarasota, FL, USA.,Department of Life Sciences, The Open University, Milton Keynes, UK.,James A. Haley Veteran's Administration, Tampa, FL, USA.,Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Maxwell Eisenbaum
- The Roskamp Institute, Sarasota, FL, USA.,Department of Life Sciences, The Open University, Milton Keynes, UK
| | - Moustafa Algamal
- The Roskamp Institute, Sarasota, FL, USA.,Department of Life Sciences, The Open University, Milton Keynes, UK
| | - Matilde Balbi
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Scott Ferguson
- The Roskamp Institute, Sarasota, FL, USA.,Department of Life Sciences, The Open University, Milton Keynes, UK
| | - Benoit Mouzon
- The Roskamp Institute, Sarasota, FL, USA.,Department of Life Sciences, The Open University, Milton Keynes, UK.,James A. Haley Veteran's Administration, Tampa, FL, USA
| | | | - Joseph Ojo
- The Roskamp Institute, Sarasota, FL, USA.,Department of Life Sciences, The Open University, Milton Keynes, UK.,James A. Haley Veteran's Administration, Tampa, FL, USA
| | - Ramon Diaz-Arrastia
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Mike Mullan
- The Roskamp Institute, Sarasota, FL, USA.,Department of Life Sciences, The Open University, Milton Keynes, UK
| | - Fiona Crawford
- The Roskamp Institute, Sarasota, FL, USA.,Department of Life Sciences, The Open University, Milton Keynes, UK.,James A. Haley Veteran's Administration, Tampa, FL, USA
| | - Corbin Bachmeier
- The Roskamp Institute, Sarasota, FL, USA.,Department of Life Sciences, The Open University, Milton Keynes, UK.,Bay Pines VA Healthcare System, Bay Pines, FL, USA
| |
Collapse
|
6
|
Prikhodko V, Chernyuk D, Sysoev Y, Zernov N, Okovityi S, Popugaeva E. Potential Drug Candidates to Treat TRPC6 Channel Deficiencies in the Pathophysiology of Alzheimer's Disease and Brain Ischemia. Cells 2020; 9:cells9112351. [PMID: 33114455 PMCID: PMC7692306 DOI: 10.3390/cells9112351] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/28/2020] [Accepted: 10/20/2020] [Indexed: 12/16/2022] Open
Abstract
Alzheimer’s disease and cerebral ischemia are among the many causative neurodegenerative diseases that lead to disabilities in the middle-aged and elderly population. There are no effective disease-preventing therapies for these pathologies. Recent in vitro and in vivo studies have revealed the TRPC6 channel to be a promising molecular target for the development of neuroprotective agents. TRPC6 channel is a non-selective cation plasma membrane channel that is permeable to Ca2+. Its Ca2+-dependent pharmacological effect is associated with the stabilization and protection of excitatory synapses. Downregulation as well as upregulation of TRPC6 channel functions have been observed in Alzheimer’s disease and brain ischemia models. Thus, in order to protect neurons from Alzheimer’s disease and cerebral ischemia, proper TRPC6 channels modulators have to be used. TRPC6 channels modulators are an emerging research field. New chemical structures modulating the activity of TRPC6 channels are being currently discovered. The recent publication of the cryo-EM structure of TRPC6 channels should speed up the discovery process even more. This review summarizes the currently available information about potential drug candidates that may be used as basic structures to develop selective, highly potent TRPC6 channel modulators to treat neurodegenerative disorders, such as Alzheimer’s disease and cerebral ischemia.
Collapse
Affiliation(s)
- Veronika Prikhodko
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia; (V.P.); (D.C.); (Y.S.); (N.Z.)
- Department of Pharmacology and Clinical Pharmacology, Saint Petersburg State Chemical Pharmaceutical University, 197022 St. Petersburg, Russia;
- N.P. Bechtereva Institute of the Human Brain of the Russian Academy of Sciences, 197376 St. Petersburg, Russia
| | - Daria Chernyuk
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia; (V.P.); (D.C.); (Y.S.); (N.Z.)
| | - Yurii Sysoev
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia; (V.P.); (D.C.); (Y.S.); (N.Z.)
- Department of Pharmacology and Clinical Pharmacology, Saint Petersburg State Chemical Pharmaceutical University, 197022 St. Petersburg, Russia;
- N.P. Bechtereva Institute of the Human Brain of the Russian Academy of Sciences, 197376 St. Petersburg, Russia
- Institute of Translational Biomedicine, Saint Petersburg State University, 199034 St. Petersburg, Russia
| | - Nikita Zernov
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia; (V.P.); (D.C.); (Y.S.); (N.Z.)
| | - Sergey Okovityi
- Department of Pharmacology and Clinical Pharmacology, Saint Petersburg State Chemical Pharmaceutical University, 197022 St. Petersburg, Russia;
- N.P. Bechtereva Institute of the Human Brain of the Russian Academy of Sciences, 197376 St. Petersburg, Russia
| | - Elena Popugaeva
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia; (V.P.); (D.C.); (Y.S.); (N.Z.)
- Correspondence:
| |
Collapse
|
7
|
Hunter LW, Jayachandran M, Miller VM. Sex differences in the expression of cell adhesion molecules on microvesicles derived from cultured human brain microvascular endothelial cells treated with inflammatory and thrombotic stimuli. Biol Sex Differ 2019; 10:26. [PMID: 31118073 PMCID: PMC6532199 DOI: 10.1186/s13293-019-0241-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 05/13/2019] [Indexed: 01/06/2023] Open
Abstract
Background There are sex differences in risk for stroke and small vessel ischemic disease in the brain. Microvesicles (MV) derived from activated cells vary by cell of origin and the stimulus initiating their release. MV released from cells activated by inflammatory and thrombotic factors have the potential to disrupt endothelial cells of the brain microvasculature. Therefore, experiments were designed to identify sex differences in the phenotype of MV released from cultured human brain microvascular endothelial cells (HBMEC) in response to inflammatory and thrombotic stimuli. Methods Cultured HBMEC derived from 20- to 30-year-old male and female donors were treated for 20 h with medium supplemented with tumor necrosis factor alpha (TNFα; 20 ng/ml), thrombin (THR; 2 U/ml), or vehicle (i.e., control). MV were isolated from the conditioned media by high-speed centrifugation and quantified by digital flow cytometry by labeling with fluorophore-conjugated primary antibodies against PECAM-1, integrin αvβ3, ICAM-1, E-selectin, or MCAM. In addition, temporal uptake of labeled MV into control HBMEC was examined by confocal microscopy. Results Under control conditions, male HBMEC released fewer MV expressing each antigen, except for PECAM-1, than female cells (P < 0.05). Neither TNFα nor THR reduced cell viability. However, TNFα induced apoptosis in female and male cells, whereas THR increased apoptosis marginally only in male cells. TNFα increased expression of all antigens tested on MV in male cells, but only increased expression of integrin αvβ3, ICAM-1, and E-selectin on MV from female cells. THR increased expression of PECAM-1, ICAM-1, and MCAM-1 on MV from male but not female cells. MV were internalized and localized to lysosomes within 90 min after their application to HBMEC. Conclusions There are sex differences in expression of cell adhesion molecules on MV released from HBMEC under control conditions and upon activation by TNFα or THR. MV taken up by unstimulated HBMEC may impact the integrity of the brain microvasculature and account, in part, for sex differences in vascular pathologies in the brain.
Collapse
Affiliation(s)
- Larry W Hunter
- Department of Surgery, Mayo Clinic, Medical Science Bldg. 4-20, 200 First St. SW, Rochester, MN, 55905, USA
| | - Muthuvel Jayachandran
- Department of Surgery, Mayo Clinic, Medical Science Bldg. 4-20, 200 First St. SW, Rochester, MN, 55905, USA.,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA.,Divisions of Hematology Research and Nephrology and Hypertension Research, Mayo Clinic, Rochester, MN, 55905, USA
| | - Virginia M Miller
- Department of Surgery, Mayo Clinic, Medical Science Bldg. 4-20, 200 First St. SW, Rochester, MN, 55905, USA. .,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA. .,Women's Health Research Center, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
8
|
Chen X, Taylor-Nguyen NN, Riley AM, Herring BP, White FA, Obukhov AG. The TRPC6 inhibitor, larixyl acetate, is effective in protecting against traumatic brain injury-induced systemic endothelial dysfunction. J Neuroinflammation 2019; 16:21. [PMID: 30704505 PMCID: PMC6354413 DOI: 10.1186/s12974-019-1407-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Accepted: 01/11/2019] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND The incidence of traumatic brain injuries (TBIs) is on the rise in the USA. Concussions, or mild TBIs without skull fracture, account for about 75% of all TBIs. Mild TBIs (mTBIs) lead to memory and cognitive deficits, headaches, intraocular pressure rises, axonal degeneration, neuroinflammation, and an array of cerebrovascular dysfunctions, including increased vascular permeability and decreased cerebral blood flow. It has been recently reported that besides vascular dysfunction in the cerebral circulation, mTBI may also cause a significant impairment of endothelial function in the systemic circulation, at least within mesenteric microvessels. In this study, we investigated whether mTBI affects endothelial function in aortas and determined the contribution of transient receptor potential canonical (TRPC) channels to modulating mTBI-associated endothelial dysfunction. METHODS We used a model of closed-head mTBI in C57BL/6, 129S, 129S-C57BL/6-F2 mice, and 129S-TRPC1 and 129S-C57BL/6-TRPC6 knockout mice to determine the effect of mTBI on endothelial function in mouse aortas employing ex vivo isometric tension measurements. Aortic tissue was also analyzed using immunofluorescence and qRT-PCR for TRPC6 expression following mTBI. RESULTS We show that in various strains of mice, mTBI induces a pronounced and long-lasting endothelial dysfunction in the aorta. Ablation of TRPC6 protects mice from mTBI-associated aortic endothelial dysfunction, while TRPC1 ablation does not impact brain injury-induced endothelial impairment in the aorta. Consistent with a role of TRPC6 activation following mTBI, we observed improved endothelial function in wild type control mice subjected to mTBI following 7-day in vivo treatment with larixyl acetate, an inhibitor of TRPC6 channels. Conversely, in vitro treatment with the pro-inflammatory endotoxin lipopolysaccharide, which activates endothelial TRPC6 in a Toll-like receptor type 4 (TLR4)-dependent manner, worsened aortic endothelial dysfunction in wild type mice. Lipopolysaccharide treatment in vitro failed to elicit endothelial dysfunction in TRPC6 knockout mice. No change in endothelial TRPC6 expression was observed 7 days following TBI. CONCLUSIONS These data suggest that TRPC6 activation may be critical for inducing endothelial dysfunction following closed-head mTBI and that pharmacological inhibition of the channel may be a feasible therapeutic strategy for preventing mTBI-associated systemic endothelial dysfunction.
Collapse
Affiliation(s)
- Xingjuan Chen
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Natalie N. Taylor-Nguyen
- Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN 46202 USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Ashley M. Riley
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - B. Paul Herring
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Fletcher A. White
- Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN 46202 USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Alexander G. Obukhov
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202 USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| |
Collapse
|
9
|
Dong B, Yang Y, Zhang Z, Xie K, Su L, Yu Y. Hemopexin alleviates cognitive dysfunction after focal cerebral ischemia-reperfusion injury in rats. BMC Anesthesiol 2019; 19:13. [PMID: 30646866 PMCID: PMC6334464 DOI: 10.1186/s12871-019-0681-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 01/04/2019] [Indexed: 01/15/2023] Open
Abstract
Background Ischemia-reperfusion (I/R) is a critical pathophysiological basis of cognitive dysfunction caused by ischemia stroke. Heme-oxygenase-1 (HO-1) is the rate-limiting enzyme for the elimination of excessive free heme by combining with hemopexin (HPX), a plasma protein that contributes to eliminating excessive free heme during ischemia stroke. This study aimed to elucidate whether HPX could alleviate cognitive dysfunction in rats subjected to cerebral I/R. Methods Rats were randomly divided into five groups: sham, MCAO, Vehicle, HPX and HPX + protoporphyrin IX (ZnPPIX). Cerebral I/R was induced by MCAO. Saline, vehicle, HPX and HPX + ZnPPIX were injected intracerebroventricularly at the moment after reperfusion. Morris water maze (MWM) test was used to detect the learning and cognitive function. Western blot was used to detect the expression of HO-1 in ischemic penumbra. CD31/vWF double labeling immunofluorescence was used to detect the neovascularization in the penumbra hippocampus. The structure and function of blood-brain barrier (BBB) was detected by the permeability of Evans Blue (EB), water content of the brain tissue, the Ang1/Ang2 and VE-cadherin expression. Results Our study verified that HPX improved the learning and memory capacity. Hemopexin up-regulated HO-1 protein expression, the average vessel density in the penumbra hippocampus and the VE- cadherin expression but decreased the permeability of EB, the water content of brain tissue and the ratio of Ang1/Ang2. The effects were reversed by ZnPPIX, an inhibitor of HO-1. Conclusion HPX can maintain the integrity of the blood-brain barrier and alleviate cognitive dysfunction after cerebral I/R through the HO-1 pathway.
Collapse
Affiliation(s)
- Beibei Dong
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052, People's Republic of China
| | - Yongyan Yang
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052, People's Republic of China
| | - Zhishen Zhang
- Department of Anesthesiology, Xiamen Medical University Zhongshan Hospital, Xiamen, 516211, China
| | - Keliang Xie
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052, People's Republic of China.
| | - Lin Su
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052, People's Republic of China
| | - Yonghao Yu
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052, People's Republic of China.
| |
Collapse
|
10
|
Kong Z, Hong Y, Zhu J, Cheng X, Liu Y. Endothelial progenitor cells improve functional recovery in focal cerebral ischemia of rat by promoting angiogenesis via VEGF. J Clin Neurosci 2018; 55:116-121. [PMID: 30041898 DOI: 10.1016/j.jocn.2018.07.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 05/13/2018] [Accepted: 07/08/2018] [Indexed: 02/05/2023]
Abstract
To investigate the role of venous infusion of endothelial progenitor cells (EPCs) in the reendothelialization of acute focal cerebral ischemia model in rats. And explore the mechanism of VEGF to promote angiogenesis of functional recovery in focal cerebral ischemia of rat. A model of middle cerebral artery occlusion (MCAo) was used to mimic ischemia following EPCs extraction from the same donor rats. EPCs were characterized by CD34, CD45 and CD133 expressions, and confirmed by uptake of fluorescently labeled Dil-ac-LDL and FITC-UEA-1 and flow cytometry analysis. EPCs were expanded in vitro and injected into the jugular vein of the same donor animals daily for 5 days after ischemia surgery. EPC-treated animals received approximately 1 × 106 cells, while control animals received PBS. Animals were evaluated the functional recovery, endothelial cell proliferation, vascular distribution, and VEGF levels. The EPC-treated group showed lower infarct volume and a significant recovery of neurological function. We also observed increased vascular distribution through bromodeoxyuridine (BrdU) staining and high plasma VEGF levels in the EPC-treated group compared to control groups. Our results provided direct evidence that auto-graft EPCs can improve neurological outcome and revascularization after ischemic stroke and indicated an important role of VEGF in this process. Our study suggested that EPCs may have potential therapeutic applications for the ischemic cerebrovascular disease.
Collapse
Affiliation(s)
- Zhaohong Kong
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yan Hong
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Jiang Zhu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Xin Cheng
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yumin Liu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| |
Collapse
|
11
|
Hosseini-Beheshti E, Grau GER. Extracellular vesicles and microvascular pathology: Decoding the active dialogue. Microcirculation 2018; 26:e12485. [PMID: 29923276 DOI: 10.1111/micc.12485] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 06/15/2018] [Indexed: 02/06/2023]
Abstract
Extracellular vesicles (EV) are a heterogeneous collection of membrane-surrounded structures released from all studied cells, under both physiological and pathological conditions. These nano-size vesicles carry complex cargoes including different classes of proteins, lipids and nucleic acids and are known to act as a communication and signalling vesicles in various cellular process. In addition to their role in development and progression of pathological disorders which make them potentially great biomarkers, EV have beneficial effects, as they take part in homeostasis. In this review we have analysed the evidence for the role of microvesicles and exosomes secreted from other cells on microvascular endothelium (EV uptake) as well as the role of endothelial-derived vesicles on their neighbouring and distant cells (EV release).
Collapse
Affiliation(s)
- Elham Hosseini-Beheshti
- Vascular Immunology Unit, Department of Pathology, School of Medical Sciences, Marie Bashir Institute and The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW, Australia
| | - Georges E R Grau
- Vascular Immunology Unit, Department of Pathology, School of Medical Sciences, Marie Bashir Institute and The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|
12
|
Jin K, Wu H, Lv T, Dai J, Zhang X, Jin Y. Ethyl pyruvate attenuates delayed experimental cerebral vasospasm following subarachnoid haemorrhage in rats: possible role of JNK pathway. RSC Adv 2018; 8:7726-7734. [PMID: 35539121 PMCID: PMC9078488 DOI: 10.1039/c7ra10801j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 02/12/2018] [Indexed: 12/21/2022] Open
Abstract
The pathophysiology of delayed cerebral vasospasm (CVS) after subarachnoid haemorrhage (SAH) is multifaceted and involves endothelial apoptosis and inflammation. Ethyl pyruvate (EP) could attenuate early brain injury following SAH via anti-inflammation and inhibition of the c-Jun N-terminal kinase (JNK) signalling pathway. However, the role of EP in the delayed CVS has yet to be determined. In this study, we examined the effect of EP on endothelial apoptosis and inflammation and explore possible signalling pathways. We found that EP could significantly attenuate the delayed CVS. Possible mechanisms include a decrease in the endothelial cell apoptosis of the basilar artery and alleviation of endothelial inflammation. The JNK signalling pathway may play an important role in the neuroprotective effects of EP on delayed CVS. The results suggest that EP may be a possible therapy for delayed CVS, and the JNK signalling pathway should be targeted for therapeutic purposes in the future. The pathophysiology of delayed cerebral vasospasm (CVS) after subarachnoid haemorrhage (SAH) is multifaceted and involves endothelial apoptosis and inflammation.![]()
Collapse
Affiliation(s)
- Ke Jin
- Department of Neurosurgery
- Renji Hospital
- Shanghai Jiaotong University
- School of Medicine
- Shanghai 200127
| | - Hui Wu
- Department of Neurosurgery
- Renji Hospital
- Shanghai Jiaotong University
- School of Medicine
- Shanghai 200127
| | - Tao Lv
- Department of Neurosurgery
- Renji Hospital
- Shanghai Jiaotong University
- School of Medicine
- Shanghai 200127
| | - Jiong Dai
- Department of Neurosurgery
- Renji Hospital
- Shanghai Jiaotong University
- School of Medicine
- Shanghai 200127
| | - Xiaohua Zhang
- Department of Neurosurgery
- Renji Hospital
- Shanghai Jiaotong University
- School of Medicine
- Shanghai 200127
| | - Yichao Jin
- Department of Neurosurgery
- Renji Hospital
- Shanghai Jiaotong University
- School of Medicine
- Shanghai 200127
| |
Collapse
|
13
|
Tydén H, Lood C, Gullstrand B, Nielsen CT, Heegaard NHH, Kahn R, Jönsen A, Bengtsson AA. Endothelial dysfunction is associated with activation of the type I interferon system and platelets in patients with systemic lupus erythematosus. RMD Open 2017; 3:e000508. [PMID: 29119007 PMCID: PMC5663269 DOI: 10.1136/rmdopen-2017-000508] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 09/17/2017] [Accepted: 09/28/2017] [Indexed: 12/17/2022] Open
Abstract
Objectives Endothelial dysfunction may be connected to cardiovascular disease (CVD) in systemic lupus erythematosus (SLE). Type I interferons (IFNs) are central in SLE pathogenesis and are suggested to induce both endothelial dysfunction and platelet activation. In this study, we investigated the interplay between endothelial dysfunction, platelets and type I IFN in SLE. Methods We enrolled 148 patients with SLE and 79 sex-matched and age-matched healthy controls (HCs). Type I IFN activity was assessed with a reporter cell assay and platelet activation by flow cytometry. Endothelial dysfunction was assessed using surrogate markers of endothelial activation, soluble vascular cell adhesion molecule-1 (sVCAM-1) and endothelial microparticles (EMPs), and finger plethysmograph to determine Reactive Hyperaemia Index (RHI). Results In patients with SLE, type I IFN activity was associated with endothelial activation, measured by high sVCAM-1 (OR 1.68, p<0.01) and elevated EMPs (OR 1.40, p=0.03). Patients with SLE with high type I IFN activity had lower RHI than HCs (OR 2.61, p=0.04), indicating endothelial dysfunction.Deposition of complement factors on platelets, a measure of platelet activation, was seen in patients with endothelial dysfunction. High levels of sVCAM-1 were associated with increased deposition of C4d (OR 4.57, p<0.01) and C1q (OR 4.10, p=0.04) on platelets. High levels of EMPs were associated with C4d deposition on platelets (OR 3.64, p=0.03). Conclusions Endothelial dysfunction was associated with activation of platelets and the type I IFN system. We suggest that an interplay between the type I IFN system, injured endothelium and activated platelets may contribute to development of CVD in SLE.
Collapse
Affiliation(s)
- Helena Tydén
- Department of Rheumatology, Clinical Sciences Lund, Lunds University, Lund, Sweden
| | - Christian Lood
- Department of Rheumatology, Clinical Sciences Lund, Lunds University, Lund, Sweden
| | - Birgitta Gullstrand
- Department of Rheumatology, Clinical Sciences Lund, Lunds University, Lund, Sweden
| | - Christoffer Tandrup Nielsen
- Department of Autoimmunology and Biomarkers, Statens Serum Institut, Copenhagen, Denmark.,Copenhagen Lupus and Vasculitis Clinic, Centre for Rheumatology and Spine Diseases, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Niels H H Heegaard
- Department of Autoimmunology and Biomarkers, Statens Serum Institut, Copenhagen, Denmark.,Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark
| | - Robin Kahn
- Department of Pediatrics, Clinical Sciences Lund, Lunds Universitey, Lund, Sweden
| | - Andreas Jönsen
- Department of Rheumatology, Clinical Sciences Lund, Lunds University, Lund, Sweden
| | - Anders A Bengtsson
- Department of Rheumatology, Clinical Sciences Lund, Lunds University, Lund, Sweden
| |
Collapse
|
14
|
Xiao B, Chai Y, Lv S, Ye M, Wu M, Xie L, Fan Y, Zhu X, Gao Z. Endothelial cell-derived exosomes protect SH-SY5Y nerve cells against ischemia/reperfusion injury. Int J Mol Med 2017; 40:1201-1209. [PMID: 28849073 PMCID: PMC5593464 DOI: 10.3892/ijmm.2017.3106] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 08/11/2017] [Indexed: 12/27/2022] Open
Abstract
Cerebral ischemia is a leading cause of death and disability. A previous study indicated that remote ischemic postconditioning (RIP) in the treatment of cerebral ischemia reduces ischemia/reperfusion (I/R) injury. However, the underlying mechanism is not well understood. In the present study, the authors hypothesized that the protective effect of RIP on neurological damage is mediated by exosomes that are released by endothelial cells in femoral arteries. To test this, right middle cerebral artery occlusion/reperfusion with RIP was performed in rats. In addition, an I/R injury cell model was tested that included human umbilical vein endothelial cells (HUVECs) and SH-SY5Y cells. Both the in vivo and in vitro models were examined for injury. Markers of exosomes (CD63, HSP70 and TSG101) were assessed by immunohistochemistry, western blot analysis and flow cytometry. Exosomes were extracted from both animal serum and HUVEC culture medium and identified by electron microscopy. They investigated the role of endothelial cell-derived exosomes in the proliferation, apoptosis, cell cycle, migration and invasion of I/R-injured SH-SY5Y cells. In addition, apoptosis-related molecules caspase-3, Bax and Bcl-2 were detected. RIP was determined to increase the number of exosomes and the expression levels of CD63, HSP70 and TSG101 in plasma, but not in brain hippocampal tissue. The size of exosomes released after I/R in HUVECs was similar to the size of exosomes released in rats subjected to RIP. Endothelial cell-derived exosomes partly suppressed the I/R-induced cell cycle arrest and apoptosis, and inhibited cell proliferation, migration and invasion in SH-SY5Y nerve cells. Endothelial cell-derived exosomes directly protect nerve cells against I/R injury, and are responsible for the protective role of RIP in I/R.
Collapse
Affiliation(s)
| | | | - Shigang Lv
- Department of Neurosurgery, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Minhua Ye
- Department of Neurosurgery, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Miaojing Wu
- Department of Neurosurgery, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Liyuan Xie
- Department of Neurosurgery, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yanghua Fan
- Department of Neurosurgery, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xingen Zhu
- Department of Neurosurgery, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Ziyun Gao
- Department of Neurosurgery, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
15
|
Hernandez-Lopez R, Chavez-Gonzalez A, Torres-Barrera P, Moreno-Lorenzana D, Lopez-DiazGuerrero N, Santiago-German D, Isordia-Salas I, Smadja D, C. Yoder M, Majluf-Cruz A, Alvarado-Moreno JA. Reduced proliferation of endothelial colony-forming cells in unprovoked venous thromboembolic disease as a consequence of endothelial dysfunction. PLoS One 2017; 12:e0183827. [PMID: 28910333 PMCID: PMC5598948 DOI: 10.1371/journal.pone.0183827] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 08/11/2017] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Venous thromboembolic disease (VTD) is a public health problem. We recently reported that endothelial colony-forming cells (ECFCs) derived from endothelial cells (EC) (ECFC-ECs) from patients with VTD have a dysfunctional state. For this study, we proposed that a dysfunctional status of these cells generates a reduction of its proliferative ability, which is also associated with senescence and reactive oxygen species (ROS). METHODS AND RESULTS Human mononuclear cells (MNCs) were obtained from peripheral blood from 40 healthy human volunteers (controls) and 50 patients with VTD matched by age (20-50 years) and sex to obtain ECFCs. We assayed their proliferative ability with plasma of patients and controls and supernatants of cultures from ECFC-ECs, senescence-associated β-galactosidase (SA-β-gal), ROS, and expression of ephrin-B2/Eph-B4 receptor. Compared with cells from controls, cells from VTD patients showed an 8-fold increase of ECFCs that emerged 1 week earlier, reduced proliferation at long term (39%) and, in passages 4 and 10, a highly senescent rate (30±1.05% vs. 91.3±15.07%, respectively) with an increase of ROS and impaired expression of ephrin-B2/Eph-4 genes. Proliferation potential of cells from VTD patients was reduced in endothelial medium [1.4±0.22 doubling population (DP)], control plasma (1.18±0.31 DP), or plasma from VTD patients (1.65±0.27 DP). CONCLUSIONS As compared with controls, ECFC-ECs from individuals with VTD have higher oxidative stress, proliferation stress, cellular senescence, and low proliferative potential. These findings suggest that patients with a history of VTD are ECFC-ECs dysfunctional that could be associated to permanent risk for new thrombotic events.
Collapse
Affiliation(s)
- Rubicel Hernandez-Lopez
- Unidad de Investigacion Medica en Trombosis, Hemostasia y Aterogenesis, Instituto Mexicano del Seguro Social, Mexico City, Mexico
- Posgrado en Biologia Experimental, Universidad Autonoma Metropolitana, Iztapalapa. Mexico City, Mexico
| | - Antonieta Chavez-Gonzalez
- Unidad de Investigacion Medica en Enfermedades Oncologicas, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Patricia Torres-Barrera
- Unidad de Investigacion Medica en Enfermedades Oncologicas, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Dafne Moreno-Lorenzana
- Unidad de Investigacion Medica en Enfermedades Oncologicas, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Norma Lopez-DiazGuerrero
- Posgrado en Biologia Experimental, Universidad Autonoma Metropolitana, Iztapalapa. Mexico City, Mexico
| | | | - Irma Isordia-Salas
- Unidad de Investigacion Medica en Trombosis, Hemostasia y Aterogenesis, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - David Smadja
- Paris Descartes University, INSERM UMR-S 1140, Faculté de Pharmacie de Paris, Paris, France
- AP-HP, Hôpital Européen Georges Pompidou, Hematology department, Paris, France
| | - Mervin C. Yoder
- Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Abraham Majluf-Cruz
- Unidad de Investigacion Medica en Trombosis, Hemostasia y Aterogenesis, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - J. Antonio Alvarado-Moreno
- Unidad de Investigacion Medica en Trombosis, Hemostasia y Aterogenesis, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| |
Collapse
|
16
|
Deng F, Wang S, Zhang L. Endothelial microparticles act as novel diagnostic and therapeutic biomarkers of circulatory hypoxia-related diseases: a literature review. J Cell Mol Med 2017; 21:1698-1710. [PMID: 28316143 PMCID: PMC5571516 DOI: 10.1111/jcmm.13125] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 01/16/2017] [Indexed: 12/28/2022] Open
Abstract
Circulatory hypoxia-related diseases (CHRDs), including acute coronary syndromes, stroke and organ transplantation, attract increased attention due to high morbidity and mortality. Mounting evidence shows that hypoxia-induced oxidative stress, coagulation, inflammation and angiogenesis play extremely important roles in the physiological and pathological processes of CHRD-related vascular endothelial injury. Interestingly, hypoxia, even hypoxia-induced oxidative stress, coagulation and inflammation can all induce release of endothelial microparticles (EMPs). EMPs, shed from activated or apoptotic endothelial cells (ECs), reflect the degree of EC damage, and elevated EMP levels are found in several CHRDs. Furthermore, EMPs, which play an important role in cell-to-cell communication and function, have confirmed pro-coagulant, proinflammatory, angiogenic and other functions, affecting pathological processes. These findings suggest that EMPs and CHRDs have a very close relationship, and EMPs may help to identify CHRD phenotypes and stratify the severity of disease, to improve risk stratification for developing CHRDs, to better define prophylactic strategies and to ameliorate prognostic characterization of patients with CHRDs. This review summarizes the known and potential roles of EMPs in the diagnosis, staging, treatment and clinical prognosis of CHRDs.
Collapse
Affiliation(s)
- Fan Deng
- Department of AnesthesiologyAffiliated Hospital of Guangdong Medical UniversityZhanjiangGuangdongChina
- Guangdong Medical UniversityZhanjiangGuangdongChina
| | - Shuang Wang
- Department of AnesthesiologyAffiliated Hospital of Guangdong Medical UniversityZhanjiangGuangdongChina
- Guangdong Medical UniversityZhanjiangGuangdongChina
| | - Liangqing Zhang
- Department of AnesthesiologyAffiliated Hospital of Guangdong Medical UniversityZhanjiangGuangdongChina
| |
Collapse
|
17
|
Gybel-Brask M, Rasmussen R, Stensballe J, Johansson PI, Ostrowski SR. Effect of delayed onset prostacyclin on markers of endothelial function and damage after subarachnoid hemorrhage. Acta Neurochir (Wien) 2017; 159:1073-1078. [PMID: 28386837 DOI: 10.1007/s00701-017-3168-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 03/27/2017] [Indexed: 11/26/2022]
Abstract
BACKGROUND Subarachnoid hemorrhage (SAH) is a neurological emergency. Delayed ischemic neurological deficit is one of the main causes of poor outcome after SAH and is probably caused, at least in part, by cerebral vasospasm. The pathophysiology of this is multifaceted, but endothelial damage and activation as well as glycocalyx damage have been implicated. Prostacyclin has been shown to protect damaged and activated endothelium and to facilitate glycocalyx repair. We investigated biomarkers of endothelial activation and damage in patients with SAH randomized to 5 days prostacyclin infusion or placebo. METHODS Patients with aneurysmal SAH managed by coiling or surgery, and a World Federation of Neurological Surgeons score between 1 and 4, and Fisher grade 3 or 4, were treated with a continuous low-dose intravenous prostacyclin infusion or placebo initiated on day 5 and discontinued on day 10 after SAH. Blood samples were drawn from the patients before, during and after prostacyclin/placebo infusion. Soluble biomarkers of endothelial cell activation (sE-selectin, sVE-cadherin) and damage (sTM), glycocalyx damage (syndecan-1) and sympathoadrenal activation (adrenaline, noradrenaline), were measured by ELISA. RESULTS Ninety patients were randomized. Prostacyclin infusion influenced neither biomarkers of sympathoadrenal activation, endothelial activation and damage nor biomarkers of endothelial glycocalyx breakdown. CONCLUSIONS We did not find any effects on markers of sympathoadrenal activation, endothelial damage and activation, or glycocalyx degradation of delayed onset prostacyclin infusion compared to placebo. Further trials investigating early onset endothelial repair using prostacyclin are warranted.
Collapse
Affiliation(s)
- Mikkel Gybel-Brask
- Section for Transfusion Medicine, Capital Region Blood Bank, Rigshospitalet, University Hospital of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark.
| | - Rune Rasmussen
- Department of Neurosurgery, Neurocenter, Rigshospitalet, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Jakob Stensballe
- Section for Transfusion Medicine, Capital Region Blood Bank, Rigshospitalet, University Hospital of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark
- Department of Anesthesia, Center of Head and Orthopedics, Rigshospitalet, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Pär I Johansson
- Section for Transfusion Medicine, Capital Region Blood Bank, Rigshospitalet, University Hospital of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark
- Department of Surgery, University of Texas Health Medical School, Houston, TX, USA
- Center for Systems Biology, School of Engineering and Natural Sciences, University of Iceland, Reykjavík, Iceland
| | - Sisse R Ostrowski
- Section for Transfusion Medicine, Capital Region Blood Bank, Rigshospitalet, University Hospital of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark
| |
Collapse
|