1
|
Palanisamy K, Karpagavalli M, Nareshkumar RN, Ramasubramanyan S, Angayarkanni N, Raman R, Chidambaram S. Adiponectin-induced activation of ERK1/2 drives fibrosis in retinal pigment epithelial cells. Hum Cell 2024; 38:8. [PMID: 39460900 DOI: 10.1007/s13577-024-01131-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024]
Abstract
Adiponectin (APN), a vasoactive cytokine produced by adipocytes, has emerged as a critical player in retinal diseases. Renowned for its antioxidant, anti-angiogenic, and anti-inflammatory properties, APN levels are closely linked to metabolic disorders, such as insulin resistance, obesity, and diabetic retinopathy (DR). Our previous work demonstrated that APN is similar in efficiency as Avastin in limiting neovascularization in retinal endothelial cells. In this study, we analyzed the effect of APN on retinal epithelial cells to understand its potential impact on eye-related pathologies. Overexpression of APN in ARPE-19 cells predominantly yielded the MMW-APN form, accompanied by increased expression of pro-fibrotic markers and decreased levels of tight junction (TJ) proteins, ZO-1, and Occludin. Further, confocal imaging revealed impaired TJ assembly and the integrity of TJ was also compromised as evidenced by the higher paracellular permeability and lower TEER. Besides, rAPN treatment in ARPE-19 cells as well triggered increased expression of pro-fibrotic markers, pro-MMP2, and enhanced cell migration and proliferation. Mechanistically, these pro-fibrotic effects were mediated by APN-induced phosphorylation of ERK1/2, causing RPE cell transdifferentiation. Furthermore, we identified that MMW-APN was the most prevalent form detected in the vitreous humor of proliferative diabetic retinopathy (PDR) patients, emphasizing the clinical relevance of our findings. Overall, our data suggest that APN, particularly its MMW form, induces epithelial-mesenchymal transition (EMT) and fibrosis in RPE cells, potentially driving the angio-fibrotic shift observed in PDR via ERK1/2 activation.
Collapse
Affiliation(s)
- Karthikka Palanisamy
- Department of Biochemistry and Cell Biology, R.S. Mehta Jain, KBIRVO, Vision Research Foundation, Chennai, India
| | | | | | - Sharada Ramasubramanyan
- Department of Biochemistry and Cell Biology, R.S. Mehta Jain, KBIRVO, Vision Research Foundation, Chennai, India
| | - Narayanasamy Angayarkanni
- Department of Biochemistry and Cell Biology, R.S. Mehta Jain, KBIRVO, Vision Research Foundation, Chennai, India
| | - Rajiv Raman
- Shri Bhagwan Mahavir Vitreoretinal Services, Sankara Nethralaya, Chennai, Tamil Nadu, India
| | | |
Collapse
|
2
|
Luo H, Zhou H, Chen Y, Sun X, Li Y, Li G, Long S, Wang S, Liang G, Chen S. Hypoxia impairs urothelial barrier function by inhibiting the expression of tight junction proteins in SV-HUC-1 cells. J Cell Mol Med 2024; 28:e18545. [PMID: 39031471 PMCID: PMC11258885 DOI: 10.1111/jcmm.18545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/25/2024] [Accepted: 07/09/2024] [Indexed: 07/22/2024] Open
Abstract
Hypoxia plays an important role in the pathological process of bladder outlet obstruction. Previous research has mostly focused on the dysfunction of bladder smooth muscle cells, which are directly related to bladder contraction. This study delves into the barrier function changes of the urothelial cells under exposure to hypoxia. Results indicated that after a 5-day culture, SV-HUC-1 formed a monolayer and/or bilayer of cell sheets, with tight junction formation, but no asymmetrical unit membrane was observed. qPCR and western blotting revealed the expression of TJ-associated proteins (occludin, claudin1 and ZO-1) was significantly decreased in the hypoxia group in a time-dependent manner. No expression changes were observed in uroplakins. When compared to normoxic groups, immunofluorescent staining revealed a reduction in the expression of TJ-associated proteins in the hypoxia group. Transepithelial electrical resistance (TEER) revealed a statistically significant decrease in resistance in the hypoxia group. Fluorescein isothiocyanate-conjugated dextran assay was inversely proportional to the results of TEER. Taken together, hypoxia down-regulates the expression of TJ-associated proteins and breaks tight junctions, thus impairing the barrier function in human urothelial cells.
Collapse
Affiliation(s)
- Huijiu Luo
- Department of UrologyAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Hui Zhou
- Department of UrologyAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Yuzhu Chen
- Department of UrologyAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Xianwu Sun
- Department of UrologyAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Yihuan Li
- Department of UrologyAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Guangjie Li
- Department of UrologyAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Shouyi Long
- Department of UrologyAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Shiyu Wang
- Department of UrologyAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Guobiao Liang
- Department of UrologyAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Shulian Chen
- Department of UrologyAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| |
Collapse
|
3
|
Palanisamy K, Chidambaram S. An In Vitro Bilayer Model of Human Primary Retinal Pigment Epithelial and Choroid Endothelial Cells for Permeability Studies. Methods Mol Biol 2024; 2711:205-223. [PMID: 37776460 DOI: 10.1007/978-1-0716-3429-5_17] [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: 10/02/2023]
Abstract
The blood-retinal barrier (BRB) present in the posterior chamber of the eye plays a major role in maintaining the proper function and integrity of the retina. Retinal pigment epithelium and choriocapillaris form the outer blood retinal barrier (oBRB), and breakdown of this barrier leads to vision-threatening diseases like macular edema, macular degeneration, and diabetic retinopathy. A simplified cell culture model of oBRB will be of great importance in elucidating the molecular mechanism of the disease progression. This chapter describes methods for primary cell isolation from donor eyes to culture human retinal pigment epithelial cells (hRPE) and choroidal endothelial cells (hCEC) and the protocol for construction of a simplified in vitro model of oBRB on fibronectin-coated Transwell inserts. Further, we explained the permeability study using FITC-dextran conjugated tracers for validating the bilayer model. The permeability experiments ensured that the system could easily be manipulated to recapitulate the pathological condition in vitro. Thus, it would be an optimal system for studying the disease mechanisms related to retinal and choroidal pathologies, for screening small molecules, and for performing drug permeability kinetics. Moreover, fundamental understanding of paracellular and transcellular trafficking of cargo in hRPE and hCEC could also be studied using this model.
Collapse
Affiliation(s)
- Karthikka Palanisamy
- R.S. Mehta Jain Department of Biochemistry and Cell Biology, Vision Research Foundation, Chennai, India
| | | |
Collapse
|
4
|
Mahmoodkhani M, Aminmansour B, Shafiei M, Hasas M, Tehrani DS. Citicoline on the Barthel Index: Severe and moderate brain injury. Indian J Pharmacol 2023; 55:223-228. [PMID: 37737074 PMCID: PMC10657618 DOI: 10.4103/ijp.ijp_570_21] [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] [Indexed: 09/23/2023] Open
Abstract
INTRODUCTION Traumatic brain injury (TBI) is a paramount factor in mortality and morbidity. The clinical trials conducted to investigate the efficacy of neuroprotective agents, such as citicoline, as a therapeutic alternative for TBI have presented divergent findings. Therefore, this study aimed to evaluate and compare citicoline's effect on the Barthel Index in patients with severe and moderate brain injury. MATERIALS AND METHODS The study is a randomized clinical trial. Patients in the case group (35 patients) were treated with citicoline and the control group (34 patients) received a placebo. Data were analyzed using SPSS 16 software. RESULTS The results showed that changes in the Glasgow Coma Scale, changes in quadriceps muscle force score, Barthel Index score changes, achieving the status without intubation, and spontaneous breathing in patients treated with citicoline were not a statistically significant difference in the two groups (P > 0.05). CONCLUSION Findings revealed that citicoline did not impact the recovery process of severe and moderate TBI patients.
Collapse
Affiliation(s)
- Mehdi Mahmoodkhani
- Department of Neurosurgery, School of Medicine, Neurosciences Research Center, Kashani Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Bahram Aminmansour
- Department of Neurosurgery, School of Medicine, Al-Zahra Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mehdi Shafiei
- Department of Neurosurgery, School of Medicine, Al-Zahra Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammadreza Hasas
- Department of Neurosurgery, Isfahan University of Medical Sciences, Isfahan, Iran
| | | |
Collapse
|
5
|
Kansakar U, Trimarco V, Mone P, Varzideh F, Lombardi A, Santulli G. Choline supplements: An update. Front Endocrinol (Lausanne) 2023; 14:1148166. [PMID: 36950691 PMCID: PMC10025538 DOI: 10.3389/fendo.2023.1148166] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 02/07/2023] [Indexed: 03/08/2023] Open
Abstract
In this comprehensive review, we examine the main preclinical and clinical investigations assessing the effects of different forms of choline supplementation currently available, including choline alfoscerate (C8H20NO6P), also known as alpha-glycerophosphocholine (α-GPC, or GPC), choline bitartrate, lecithin, and citicoline, which are cholinergic compounds and precursors of acetylcholine. Extensively used as food supplements, they have been shown to represent an effective strategy for boosting memory and enhancing cognitive function.
Collapse
Affiliation(s)
- Urna Kansakar
- Department of Medicine, Division of Cardiology, Einstein Institute for Aging Research, Montefiore Health System, New York, NY, United States
| | | | - Pasquale Mone
- Department of Medicine, Division of Cardiology, Einstein Institute for Aging Research, Montefiore Health System, New York, NY, United States
- ASL Avellino, Montefiore Health System, New York, NY, United States
| | - Fahimeh Varzideh
- Department of Medicine, Division of Cardiology, Einstein Institute for Aging Research, Montefiore Health System, New York, NY, United States
| | - Angela Lombardi
- Department of Microbiology and Immunology, Montefiore Health System, New York, NY, United States
- *Correspondence: Angela Lombardi,
| | - Gaetano Santulli
- Department of Medicine, Division of Cardiology, Einstein Institute for Aging Research, Montefiore Health System, New York, NY, United States
- University of Naples “Federico II”, Naples, Italy
- Department of Molecular Pharmacology, Einstein-Sinai Diabetes Research Center (ES-DRC), Montefiore Health System, New York, NY, United States
| |
Collapse
|
6
|
Al-kuraishy HM, Al-Buhadily AK, Al-Gareeb AI, Alorabi M, Hadi Al-Harcan NA, El-Bouseary MM, Batiha GES. Citicoline and COVID-19: vis-à-vis conjectured. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2022; 395:1463-1475. [PMID: 36063198 PMCID: PMC9442587 DOI: 10.1007/s00210-022-02284-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/23/2022] [Indexed: 11/29/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is a current pandemic disease caused by a novel severe acute respiratory syndrome coronavirus virus respiratory type 2 (SARS-CoV-2). SARS-CoV-2 infection is linked with various neurological manifestations due to cytokine-induced disruption of the blood brain barrier (BBB), neuroinflammation, and peripheral neuronal injury, or due to direct SARS-CoV-2 neurotropism. Of note, many repurposed agents were included in different therapeutic protocols in the management of COVID-19. These agents did not produce an effective therapeutic eradication of SARS-CoV-2, and continuing searching for novel anti-SARS-CoV-2 agents is a type of challenge nowadays. Therefore, this study aimed to review the potential anti-inflammatory and antioxidant effects of citicoline in the management of COVID-19.
Collapse
Affiliation(s)
- Hayder M. Al-kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriya University, Baghdad, Iraq
| | - Ali K. Al-Buhadily
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriya University, Baghdad, Iraq
| | - Ali I. Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriya University, Baghdad, Iraq
| | - Mohammed Alorabi
- Department of Biotechnology, College of Sciences, Taif University, P.O. Box 11099, Taif, 21944 Saudi Arabia
| | - Nasser A. Hadi Al-Harcan
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Rasheed University College, Baghdad, Iraq
| | - Maisra M. El-Bouseary
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, AlBeheira, Damanhour, 22511 Egypt
| |
Collapse
|
7
|
Abdi Sarabi M, Shiri A, Aghapour M, Reichardt C, Brandt S, Mertens PR, Medunjanin S, Bruder D, Braun-Dullaeus RC, Weinert S. Normoxic HIF-1α Stabilization Caused by Local Inflammatory Factors and Its Consequences in Human Coronary Artery Endothelial Cells. Cells 2022; 11:cells11233878. [PMID: 36497143 PMCID: PMC9737288 DOI: 10.3390/cells11233878] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
Abstract
Knowledge about normoxic hypoxia-inducible factor (HIF)-1α stabilization is limited. We investigated normoxic HIF-1α stabilization and its consequences using live cell imaging, immunoblotting, Bio-Plex multiplex immunoassay, immunofluorescence staining, and barrier integrity assays. We demonstrate for the first time that IL-8 and M-CSF caused HIF-1α stabilization and translocation into the nucleus under normoxic conditions in both human coronary endothelial cells (HCAECs) and HIF-1α-mKate2-expressing HEK-293 cells. In line with the current literature, our data show significant normoxic HIF-1α stabilization caused by TNF-α, INF-γ, IL-1β, and IGF-I in both cell lines, as well. Treatment with a cocktail consisting of TNF-α, INF-γ, and IL-1β caused significantly stronger HIF-1α stabilization in comparison to single treatments. Interestingly, this cumulative effect was not observed during simultaneous treatment with IL-8, M-CSF, and IGF-I. Furthermore, we identified two different kinetics of HIF-1α stabilization under normoxic conditions. Our data demonstrate elevated protein levels of HIF-1α-related genes known to be involved in the development of atherosclerosis. Moreover, we demonstrate an endothelial barrier dysfunction in HCAECs upon our treatments and during normoxic HIF-1α stabilization comparable to that under hypoxia. This study expands the knowledge of normoxic HIF-1α stabilization and activation and its consequences on the endothelial secretome and barrier function. Our data imply an active role of HIF-1α in vivo in the vasculature in the absence of hypoxia.
Collapse
Affiliation(s)
- Mohsen Abdi Sarabi
- Department of Internal Medicine, Division of Cardiology and Angiology, Otto-von-Guericke University, 39120 Magdeburg, Germany
| | - Alireza Shiri
- Department of Internal Medicine, Division of Cardiology and Angiology, Otto-von-Guericke University, 39120 Magdeburg, Germany
| | - Mahyar Aghapour
- Department of Internal Medicine, Division of Cardiology and Angiology, Otto-von-Guericke University, 39120 Magdeburg, Germany
- Infection Immunology Group, Institute of Medical Microbiology and Hospital Hygiene, Otto-von-Guericke University, 39120 Magdeburg, Germany
| | - Charlotte Reichardt
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University, 39120 Magdeburg, Germany
| | - Sabine Brandt
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University, 39120 Magdeburg, Germany
| | - Peter R. Mertens
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University, 39120 Magdeburg, Germany
| | - Senad Medunjanin
- Department of Internal Medicine, Division of Cardiology and Angiology, Otto-von-Guericke University, 39120 Magdeburg, Germany
| | - Dunja Bruder
- Infection Immunology Group, Institute of Medical Microbiology and Hospital Hygiene, Otto-von-Guericke University, 39120 Magdeburg, Germany
- Immune Regulation Group, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Ruediger C. Braun-Dullaeus
- Department of Internal Medicine, Division of Cardiology and Angiology, Otto-von-Guericke University, 39120 Magdeburg, Germany
- Correspondence: (R.C.B.-D.); (S.W.)
| | - Sönke Weinert
- Department of Internal Medicine, Division of Cardiology and Angiology, Otto-von-Guericke University, 39120 Magdeburg, Germany
- Correspondence: (R.C.B.-D.); (S.W.)
| |
Collapse
|
8
|
Abstract
This review is based on the previous one published in 2016 (Secades JJ. Citicoline: pharmacological and clinical review, 2016 update. Rev Neurol 2016; 63 (Supl 3): S1-S73), incorporating 176 new references, having all the information available in the same document to facilitate the access to the information in one document. This review is focused on the main indications of the drug, as acute stroke and its sequelae, including the cognitive impairment, and traumatic brain injury and its sequelae. There are retrieved the most important experimental and clinical data in both indications.
Collapse
Affiliation(s)
- Julio J. Secades
- Departamento Médico. Grupo Ferrer, S.A. Barcelona, EspañaDepartamento MédicoDepartamento MédicoBarcelonaEspaña
| | - Pietro Gareri
- Center for Cognitive Disorders and Dementia - Catanzaro Lido. ASP Catanzaro. Catanzaro, ItaliaCenter for Cognitive Disorders and Dementia - Catanzaro LidoCenter for Cognitive Disorders and Dementia - Catanzaro LidoCatanzaroItalia
| |
Collapse
|
9
|
Misan N, Michalak S, Rzymski P, Poniedziałek B, Kapska K, Osztynowicz K, Ropacka-Lesiak M. Molecular Indicators of Blood-Brain Barrier Breakdown and Neuronal Injury in Pregnancy Complicated by Fetal Growth Restriction. Int J Mol Sci 2022; 23:ijms232213798. [PMID: 36430274 PMCID: PMC9695431 DOI: 10.3390/ijms232213798] [Citation(s) in RCA: 6] [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: 09/22/2022] [Revised: 11/02/2022] [Accepted: 11/02/2022] [Indexed: 11/11/2022] Open
Abstract
This study evaluated the damage to the endothelial tight junctions (TJs) in pregnancies complicated by fetal growth restriction (FGR) and investigated whether FGR is related to blood-brain barrier disintegration and, subsequently, to the appearance of proteins indicative of neuronal injury in maternal blood. The studied group included 90 pregnant women diagnosed with FGR. The control group consisted of 70 women with an uncomplicated pregnancy. The biochemical measurements included serum neuronal proteins (subunit of the N-methyl-D-aspartate receptor-NR1, nucleoside diphosphate kinase A-NME1, and S100 calcium-binding protein B-S100B), serum TJ proteins (occludin-OCLN, claudin-5-CLN5, zonula occludens-zo-1, and OCLN/zo-1 and CLN5/zo-1 ratios), and placental expression of TJ proteins (OCLN, claudin-4 CLN4, CLN5, zo-1). The significantly higher serum S100B and CLN5 levels and serum CLN5/zo-1 ratio were observed in FGR compared to healthy pregnancies. Moreover, FGR was characterized by increased placental CLN5 expression. Both serum NME1 levels and placental CLN4 expression in FGR pregnancies were significantly related to the incidence of neurological disorders in newborns. Mothers of FGR neonates who developed neurological complications and intraventricular hemorrhage (IVH) had statistically higher NME1 concentrations during pregnancy and significantly lower placental CLN4 expression than mothers of FGR neonates without neurological abnormalities. The serum NME1 levels and placental CLN4 expression were predictive markers of IVH in the FGR group. The blood-brain barrier is destabilized in pregnancies complicated by FGR. Neurological disorders, including IVH, are associated with higher serum concentrations of NME1 and the decreased placental expression of CLN4. The serum NME1 levels and placental CLN4 expression may serve as biomarkers, helpful in predicting IVH in FGR. It may allow for more precise monitoring and influence decision-making on the optimal delivery time to avoid developing neurological complications.
Collapse
Affiliation(s)
- Natalia Misan
- Department of Perinatology and Gynecology, Poznan University of Medical Sciences, 60-535 Poznan, Poland
- Correspondence:
| | - Sławomir Michalak
- Department of Neurochemistry and Neuropathology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Piotr Rzymski
- Department of Environmental Medicine, Poznan University of Medical Sciences, 61-848 Poznan, Poland
- Integrated Science Association (ISA), Universal Scientific Education and Research Network (USERN), 60-806 Poznań, Poland
| | - Barbara Poniedziałek
- Department of Environmental Medicine, Poznan University of Medical Sciences, 61-848 Poznan, Poland
| | - Katarzyna Kapska
- Department of Perinatology and Gynecology, Poznan University of Medical Sciences, 60-535 Poznan, Poland
| | - Krystyna Osztynowicz
- Department of Neurochemistry and Neuropathology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Mariola Ropacka-Lesiak
- Department of Perinatology and Gynecology, Poznan University of Medical Sciences, 60-535 Poznan, Poland
| |
Collapse
|
10
|
Roy P, Tomassoni D, Nittari G, Traini E, Amenta F. Effects of choline containing phospholipids on the neurovascular unit: A review. Front Cell Neurosci 2022; 16:988759. [PMID: 36212684 PMCID: PMC9541750 DOI: 10.3389/fncel.2022.988759] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
The roles of choline and of choline-containing phospholipids (CCPLs) on the maintenance and progress of neurovascular unit (NVU) integrity are analyzed. NVU is composed of neurons, glial and vascular cells ensuring the correct homeostasis of the blood-brain barrier (BBB) and indirectly the function of the central nervous system. The CCPLs phosphatidylcholine (lecithin), cytidine 5′-diphosphocholine (CDP-choline), choline alphoscerate or α-glyceryl-phosphorylcholine (α-GPC) contribute to the modulation of the physiology of the NVU cells. A loss of CCPLs contributes to the development of neurodegenerative diseases such as Alzheimer’s disease, multiple sclerosis, Parkinson’s disease. Our study has characterized the cellular components of the NVU and has reviewed the effect of lecithin, of CDP-choline and α-GPC documented in preclinical studies and in limited clinical trials on these compounds. The interesting results obtained with some CCPLs, in particular with α-GPC, probably would justify reconsideration of the most promising molecules in larger attentively controlled studies. This can also contribute to better define the role of the NVU in the pathophysiology of brain disorders characterized by vascular impairment.
Collapse
Affiliation(s)
- Proshanta Roy
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Daniele Tomassoni
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Giulio Nittari
- School of Medicinal and Health Products Sciences, University of Camerino, Camerino, Italy
| | - Enea Traini
- School of Medicinal and Health Products Sciences, University of Camerino, Camerino, Italy
| | - Francesco Amenta
- School of Medicinal and Health Products Sciences, University of Camerino, Camerino, Italy
- *Correspondence: Francesco Amenta,
| |
Collapse
|
11
|
Yong KW, Janmaleki M, Pachenari M, Mitha AP, Sanati-Nezhad A, Sen A. Engineering a 3D human intracranial aneurysm model using liquid-assisted injection molding and tuned hydrogels. Acta Biomater 2021; 136:266-278. [PMID: 34547516 DOI: 10.1016/j.actbio.2021.09.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 09/14/2021] [Accepted: 09/14/2021] [Indexed: 12/30/2022]
Abstract
Physiologically relevant intracranial aneurysm (IA) models are crucially required to facilitate testing treatment options for IA. Herein, we report the development of a new in vitro tissue-engineered platform, which recapitulates the microenvironment, structure, and cellular complexity of native human IA. A new modified liquid-assisted injection molding technique was developed to fabricate a three-dimensional hollow IA model with clinically relevant IA dimensions within a mechanically tuned Gelatin Methacryloyl (GelMA) hydrogel. An endothelium lining was created inside the IA model by culturing human umbilical vein endothelial cells over pre-cultured human brain vascular smooth muscle cells. These cellularized IA models were subjected to medium perfusion at flow rates between 6.3 and 15.75 mL/min for inducing biomimetic vessel wall shear stress (10-25 dyn/cm2) to the cells for ten days. Both cell types maintained their secretome profiles and showed more than 96% viability, demonstrating the biocompatibility of the hydrogel during perfusion cell culture at such flow rates. Based on the characterized viscoelastic properties of the GelMA hydrogel and with the aid of a fluid-structure interaction model, the capability of the IA model in predicting the response of the IA to different fluid flow profiles was mathematically shown. With physiologically relevant behavior, our developed in vitro human IA model could allow researchers to better understand the pathophysiology and treatment of IA. STATEMENT OF SIGNIFICANCE: A three-dimensional intracranial aneurysm (IA) tissue model recapitulating the microenvironment, structure, and cellular complexity of native human IA was developed. • An endothelium lining was created inside the IA model over pre-cultured human brain vascular smooth muscle cells over at least 10-day successful culture. • The cells maintained their secretome profiles, demonstrating the biocompatibility of hydrogel during a long-term perfusion cell culture. • The IA model showed its capability in predicting the response of IA to different fluid flow profiles. • The cells in the vessel region behaved differently from cells in the aneurysm region due to alteration in hemodynamic shear stress. • The IA model could allow researchers to better understand the pathophysiology and treatment options of IA.
Collapse
|
12
|
High-throughput sequencing revealed the expression profile and potential key molecules of the circular RNAs involved in the process of hypoxic adaptation in Tibetan chickens. Biologia (Bratisl) 2021. [DOI: 10.1007/s11756-021-00848-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
13
|
Inada M, Xu H, Takeuchi M, Ito M, Chen M. Microglia increase tight-junction permeability in coordination with Müller cells under hypoxic condition in an in vitro model of inner blood-retinal barrier. Exp Eye Res 2021; 205:108490. [PMID: 33607076 DOI: 10.1016/j.exer.2021.108490] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 01/24/2021] [Accepted: 02/05/2021] [Indexed: 01/20/2023]
Abstract
Microglia and Müller cells (MCs) are believed to be critically involved in hypoxia-induced blood-retinal barrier (BRB) disruption, which is a major pathogenic factor of various retinopathies. However, the underlying mechanism remains poorly defined. The inner BRB (iBRB) is primarily formed of microvascular endothelial cells (ECs) with tight junction (TJ), which are surrounded and supported by retinal glial cells. We developed a novel in vitro iBRB model sheet by sandwiching Transwell membrane with layered mouse brain microvascular ECs (bEnd.3) and mouse retinal MCs (QMMuC-1) on each side of the membrane. Using this model, we tested the hypothesis that under hypoxic condition, activated microglia produce inflammatory cytokines such as interleukin (IL)-1β, which may promote vascular endothelial growth factor (VEGF) production from MCs, leading to TJ disruption. The iBRB model cell sheets were exposed to 1% oxygen for 6 h with or without mouse brain microglia (BV2) or IL-1β. TJ structure and function were examined by zonula occludens (ZO)-1 immunostaining and fluorescein isothiocyanate permeability assay, respectively. Relative gene expression of IL-1β in BV2 under normoxic and hypoxic conditions was examined by real-time reverse transcription-polymerase chain reaction. VEGF protein concentration in QMMuC-1 supernatants was measured by enzyme-linked immunosorbent assay. The bEnd.3 cell sheet incubated with BV2 in hypoxic condition or with IL-1β in normoxic condition showed abnormal localization of ZO-1 and aberrated barrier function. Under normoxic condition, EC-MC iBRB model cell sheet showed lower permeability than bEnd.3 cell sheet. Under hypoxic conditions, the barrier function of EC-MC iBRB model cell sheet was more deteriorated compared to bEnd.3 cell sheet. Under hypoxic condition, incubation of EC-MC iBRB model cell sheet with BV2 cells or IL-1β significantly increased barrier permeability, and hypoxia-treated BV2 cells expressed significantly higher levels of IL-1β mRNA. Incubation of QMMuC-1 with IL-1β increased VEGF production. These results suggest that under hypoxic condition, microglia are activated to release proinflammatory cytokines such as IL-1β that promote VEGF production from MCs, leading to disruption of iBRB function. Modulating microglia and MCs function may be a novel approach to treat hypoxia-induced retinal BRB dysfunction.
Collapse
Affiliation(s)
- Makoto Inada
- Department of Ophthalmology, National Defense Medical College, Namiki 3-2, Tokorozawa, Saitama, 359-0042, Japan; Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, Northern Ireland, United Kingdom
| | - Heping Xu
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, Northern Ireland, United Kingdom
| | - Masaru Takeuchi
- Department of Ophthalmology, National Defense Medical College, Namiki 3-2, Tokorozawa, Saitama, 359-0042, Japan
| | - Masataka Ito
- Department of Developmental Anatomy, National Defense Medical College, Namiki 3-2, Tokorozawa, Saitama, 359-0042, Japan.
| | - Mei Chen
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, Northern Ireland, United Kingdom.
| |
Collapse
|
14
|
miR-132-3p priming enhances the effects of mesenchymal stromal cell-derived exosomes on ameliorating brain ischemic injury. Stem Cell Res Ther 2020; 11:260. [PMID: 32600449 PMCID: PMC7322840 DOI: 10.1186/s13287-020-01761-0] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 05/28/2020] [Accepted: 06/08/2020] [Indexed: 02/07/2023] Open
Abstract
Backgrounds/aims Mesenchymal stromal cell-derived exosomes (MSC-EXs) could exert protective effects on recipient cells by transferring the contained microRNAs (miRs), and miR-132-3p is one of angiogenic miRs. However, whether the combination of MSC-EXs and miR-132-3p has better effects in ischemic cerebrovascular disease remains unknown. Methods Mouse MSCs transfected with scrambler control or miR-132-3p mimics were used to generate MSC-EXs and miR-132-3p-overexpressed MSC-EXs (MSC-EXsmiR-132-3p). The effects of EXs on hypoxia/reoxygenation (H/R)-injured ECs in ROS generation, apoptosis, and barrier function were analyzed. The levels of RASA1, Ras, phosphorylations of PI3K, Akt and endothelial nitric oxide synthesis (eNOS), and tight junction proteins (Claudin-5 and ZO-1) were measured. Ras and PI3K inhibitors were used for pathway analysis. In transient middle cerebral artery occlusion (tMCAO) mouse model, the effects of MSC-EXs on the cerebral vascular ROS production and apoptosis, cerebral vascular density (cMVD), Evans blue extravasation, brain water content, neurological deficit score (NDS), and infarct volume were determined. Results MSC-EXs could deliver their carried miR-132-3p into target ECs, which functionally downregulated the target protein RASA1, while upregulated the expression of Ras and the downstream PI3K phosphorylation. Compared to MSC-EXs, MSC-EXsmiR-132-3p were more effective in decreasing ROS production, apoptosis, and tight junction disruption in H/R-injured ECs. These effects were associated with increased levels of phosphorylated Akt and eNOS, which could be abolished by PI3K inhibitor (LY294002) or Ras inhibitor (NSC 23766). In the tMCAO mouse model, the infusion of MSC-EXsmiR-132-3p was more effective than MSC-EXs in reducing cerebral vascular ROS production, BBB dysfunction, and brain injury. Conclusion Our results suggest that miR-132-3p promotes the beneficial effects of MSC-EXs on brain ischemic injury through protecting cerebral EC functions.
Collapse
|
15
|
Zhang H, Pan Q, Xie Z, Chen Y, Wang J, Bihl J, Zhong W, Chen Y, Zhao B, Ma X. Implication of MicroRNA503 in Brain Endothelial Cell Function and Ischemic Stroke. Transl Stroke Res 2020; 11:1148-1164. [PMID: 32285355 DOI: 10.1007/s12975-020-00794-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 02/18/2020] [Accepted: 02/20/2020] [Indexed: 12/12/2022]
Abstract
The role of miR-503 in brain endothelium and ischemic stroke (IS) remains unclear. We aimed to study the relationship between plasma miR-503 and the onset time, severity, subtypes, and von Willebrand Factor (vWF) level in IS patients and to investigate the roles and underlying mechanisms of miR-503 in middle cerebral artery occlusion (MCAO) mice and cultured cerebral vascular endothelial cells (ECs). In MCAO mice, the effects of plasma from acute severe IS patients (ASS) with or without miR-503 antagomir on brain and ECs damage were determined. In cultured human ECs, the effects of miR-503 overexpression or knockdown on the monolayer permeability, apoptosis, ROS, and NO generation were investigated. For mechanism study, the PI3K/Akt/eNOS pathway, cleaved caspase-3, and bcl-2 were analyzed. Results showed that plasma miR-503 was significantly increased in IS patients, especially in acute period and severe cases and subtypes of LAA and TACI, and was positively correlated with vWF. Logistic analysis indicated that miR-503 was an independent risk factor for IS, with the area under curve of 0.796 in ROC analysis. In MCAO mice, ASS pretreatment aggravated neurological injury, BBB damage, brain edema, CBF reduction, and decreased NO production while increased apoptosis and ROS generation in brain ECs, which were partly abolished by miR-503 antagomir. In cultured ECs, miR-503 overexpression and knockdown confirmed its effects on regulating monolayer permeability, cell apoptosis, NO, and ROS generation via PI3K/Akt/eNOS pathway or bcl-2 and cleaved caspase-3 proteins. These together indicate that miR-503 is a promising biomarker and novel therapeutic target for IS.
Collapse
Affiliation(s)
- Huiting Zhang
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, 57 South Renmin Road, Zhanjiang, 524001, China
| | - Qunwen Pan
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, 57 South Renmin Road, Zhanjiang, 524001, China
| | - Zi Xie
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, 57 South Renmin Road, Zhanjiang, 524001, China
| | - Yanyu Chen
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, 57 South Renmin Road, Zhanjiang, 524001, China
| | - Jinju Wang
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, OH, 45430, USA
| | - Ji Bihl
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, OH, 45430, USA
| | - Wangtao Zhong
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, 57 South Renmin Road, Zhanjiang, 524001, China
| | - Yanfang Chen
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, OH, 45430, USA
| | - Bin Zhao
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, 57 South Renmin Road, Zhanjiang, 524001, China.
| | - Xiaotang Ma
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, 57 South Renmin Road, Zhanjiang, 524001, China.
| |
Collapse
|
16
|
Konijnenberg LSF, Damman P, Duncker DJ, Kloner RA, Nijveldt R, van Geuns RJM, Berry C, Riksen NP, Escaned J, van Royen N. Pathophysiology and diagnosis of coronary microvascular dysfunction in ST-elevation myocardial infarction. Cardiovasc Res 2020; 116:787-805. [PMID: 31710673 PMCID: PMC7061278 DOI: 10.1093/cvr/cvz301] [Citation(s) in RCA: 144] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 10/13/2019] [Accepted: 11/06/2019] [Indexed: 12/15/2022] Open
Abstract
Early mechanical reperfusion of the epicardial coronary artery by primary percutaneous coronary intervention (PCI) is the guideline-recommended treatment for ST-elevation myocardial infarction (STEMI). Successful restoration of epicardial coronary blood flow can be achieved in over 95% of PCI procedures. However, despite angiographically complete epicardial coronary artery patency, in about half of the patients perfusion to the distal coronary microvasculature is not fully restored, which is associated with increased morbidity and mortality. The exact pathophysiological mechanism of post-ischaemic coronary microvascular dysfunction (CMD) is still debated. Therefore, the current review discusses invasive and non-invasive techniques for the diagnosis and quantification of CMD in STEMI in the clinical setting as well as results from experimental in vitro and in vivo models focusing on ischaemic-, reperfusion-, and inflammatory damage to the coronary microvascular endothelial cells. Finally, we discuss future opportunities to prevent or treat CMD in STEMI patients.
Collapse
Affiliation(s)
- Lara S F Konijnenberg
- Department of Cardiology, Radboud University Medical Center, Postbus 9101, 6500 HB Nijmegen, The Netherlands
| | - Peter Damman
- Department of Cardiology, Radboud University Medical Center, Postbus 9101, 6500 HB Nijmegen, The Netherlands
| | - Dirk J Duncker
- Department of Radiology and Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Robert A Kloner
- Huntington Medical Research Institutes, Pasadena, CA, USA
- Division of Cardiovascular Medicine, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Robin Nijveldt
- Department of Cardiology, Radboud University Medical Center, Postbus 9101, 6500 HB Nijmegen, The Netherlands
| | - Robert-Jan M van Geuns
- Department of Cardiology, Radboud University Medical Center, Postbus 9101, 6500 HB Nijmegen, The Netherlands
| | - Colin Berry
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Clydebank, UK
- British Heart Foundation, Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Niels P Riksen
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Javier Escaned
- Department of Cardiology, Hospital Clínico San Carlos IDISSC, Universidad Complutense de Madrid, Madrid, Spain
| | - Niels van Royen
- Department of Cardiology, Radboud University Medical Center, Postbus 9101, 6500 HB Nijmegen, The Netherlands
| |
Collapse
|
17
|
Beard RS, Hoettels BA, Meegan JE, Wertz TS, Cha BJ, Yang X, Oxford JT, Wu MH, Yuan SY. AKT2 maintains brain endothelial claudin-5 expression and selective activation of IR/AKT2/FOXO1-signaling reverses barrier dysfunction. J Cereb Blood Flow Metab 2020; 40:374-391. [PMID: 30574832 PMCID: PMC7370624 DOI: 10.1177/0271678x18817512] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 10/26/2018] [Accepted: 11/14/2018] [Indexed: 12/28/2022]
Abstract
Inflammation-induced blood-brain barrier (BBB) dysfunction and microvascular leakage are associated with a host of neurological disorders. The tight junction protein claudin-5 (CLDN5) is a crucial protein necessary for BBB integrity and maintenance. CLDN5 is negatively regulated by the transcriptional repressor FOXO1, whose activity increases during impaired insulin/AKT signaling. Owing to an incomplete understanding of the mechanisms that regulate CLDN5 expression in BBB maintenance and dysfunction, therapeutic interventions remain underdeveloped. Here, we show a novel isoform-specific function for AKT2 in maintenance of BBB integrity. We identified that AKT2 during homeostasis specifically regulates CLDN5-dependent barrier integrity in brain microvascular endothelial cells (BMVECs) and that intervention with a selective insulin-receptor (IR) agonist, demethylasterriquinone B1 (DMAQ-B1), rescued IL-1β-induced AKT2 inactivation, FOXO1 nuclear accumulation, and loss of CLDN5-dependent barrier integrity. Moreover, DMAQ-B1 attenuated preclinical CLDN5-dependent BBB dysfunction in mice subjected to experimental autoimmune encephalomyelitis. Taken together, the data suggest a regulatory role for IR/AKT2/FOXO1-signaling in CLDN5 expression and BBB integrity during neuroinflammation.
Collapse
Affiliation(s)
- Richard S Beard
- Department of Molecular Pharmacology and
Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
- Department of Biological Sciences and
Biomolecular Research Center, Boise State University, Boise, ID, USA
| | - Brian A Hoettels
- Department of Biological Sciences and
Biomolecular Research Center, Boise State University, Boise, ID, USA
| | - Jamie E Meegan
- Department of Molecular Pharmacology and
Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Travis S Wertz
- Department of Biological Sciences and
Biomolecular Research Center, Boise State University, Boise, ID, USA
| | - Byeong J Cha
- Department of Molecular Pharmacology and
Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Xiaoyuan Yang
- Department of Molecular Pharmacology and
Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Julia T Oxford
- Department of Biological Sciences and
Biomolecular Research Center, Boise State University, Boise, ID, USA
| | - Mack H Wu
- Department of Surgery, Morsani College of
Medicine, University of South Florida, Tampa, FL, USA
| | - Sarah Y Yuan
- Department of Molecular Pharmacology and
Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
- Department of Surgery, Morsani College of
Medicine, University of South Florida, Tampa, FL, USA
| |
Collapse
|
18
|
Huang JH, Xu Y, Yin XM, Lin FY. Exosomes Derived from miR-126-modified MSCs Promote Angiogenesis and Neurogenesis and Attenuate Apoptosis after Spinal Cord Injury in Rats. Neuroscience 2020; 424:133-145. [PMID: 31704348 DOI: 10.1016/j.neuroscience.2019.10.043] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 10/24/2019] [Accepted: 10/25/2019] [Indexed: 12/13/2022]
Abstract
Spinal cord injury (SCI) is a devastating neurological event that results in incomplete or complete loss of voluntary motor and sensory function. Until recently, there has been no effective curative strategy for SCI. Our previous study showed that microRNA (miR)-126 promoted angiogenesis and attenuated inflammation after SCI; however, the effect of miR-126-based treatment is limited because of the low efficiency of miR delivery in vivo. Recently, accumulating evidence has indicated that exosomes can serve as a valuable therapeutic vehicle for miR delivery to the central nervous system (CNS). Thus, the present study aimed to investigate whether exosomes derived from mesenchymal stem cells (MSCs) can be used to deliver miR-126 to treat SCI. In this study, we found that MSCs can load miR-126 into secreted exosomes. In a rat model of SCI, exosomes transferred miR-126 to the injured site of the spinal cord, reduced the lesion volume and improved functional recovery after SCI. Additionally, miR-126-loaded exosomes promoted angiogenesis post-SCI. Moreover, the administration of miR-126 exosomes promoted neurogenesis and reduced cell apoptosis after SCI. In vitro, we observed that exosomes derived from miR-126-modified MSCs promoted the angiogenesis and migration of human umbilical venous endothelial cells (HUVECs) by inhibiting the expression of Sprouty-related EVH1 domain-containing protein 1 (SPRED1) and phosphoinositide-3-kinase regulatory subunit 2 (PIK3R2). In conclusion, our study demonstrated that exosomes derived from MSCs transfected with miR-126 may promote angiogenesis and neurogenesis, inhibit apoptosis and promote functional recovery after SCI. These findings suggest that exosomes derived from miR-126-modified MSCs may serve as a novel potential therapeutic strategy for treating SCI.
Collapse
Affiliation(s)
- Jiang-Hu Huang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, PR China; Department of Orthopedics, Fujian Provincial Hospital, Fujian Medical University, Fuzhou 350001, PR China
| | - Yang Xu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, PR China; Department of Orthopedics, Fujian Provincial Hospital, Fujian Medical University, Fuzhou 350001, PR China
| | - Xiao-Ming Yin
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, PR China; Department of Orthopedics, Fujian Provincial Hospital, Fujian Medical University, Fuzhou 350001, PR China
| | - Fei-Yue Lin
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, PR China; Department of Orthopedics, Fujian Provincial Hospital, Fujian Medical University, Fuzhou 350001, PR China.
| |
Collapse
|
19
|
DL-3-n-butylphthalide protects the blood-brain barrier against ischemia/hypoxia injury via upregulation of tight junction proteins. Chin Med J (Engl) 2019; 132:1344-1353. [PMID: 30939485 PMCID: PMC6629356 DOI: 10.1097/cm9.0000000000000232] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The increased permeability of the blood-brain barrier (BBB) induced by ischemia/hypoxia is generally correlated with alteration of tight junctions (TJs). DL-3-n-butylphthalide (NBP) has been shown to exert neuroprotective effects after ischemic injury. However, few studies have assessed the correlation between NBP and TJs. This study aimed to investigate the potential effect of NBP on the TJ proteins claudin-5, zonula occludens-1 (ZO-1), and occludin during brain ischemia. METHODS A chronic cerebral hypoperfusion (CCH) Sprague-Dawley rat model was established, and NBP (20, 40, or 80 mg/kg, gavage, once a day) treatment was performed for 14 days. NBP (0.1 or 1.0 μmol/L) pre-treatment was applied to an in vitro hypoxia microvascular endothelial cell model (1% O2, 24 h). BBB permeability was assessed by performing the Evans blue assay. The expressions and localization of claudin-5, ZO-1, occludin, phosphorylated/total protein kinase B (p-Akt/Akt), phosphorylated/total glycogen synthase kinase 3β (GSK-3β)/GSK-3β, and β-catenin/β-actin were evaluated by Western blotting or immunofluorescence. Reactive oxygen species (ROS) generation was measured by flow cytometry analysis. TJ ultrastructure was observed by transmission electron microscopy. RESULTS In CCH rats, treatment with 40 and 80 mg/kg NBP decreased the Evans blue content in brain tissue (9.0 ± 0.9 μg/g vs. 12.3 ± 1.9 μg/g, P = 0.005; 6.7 ± 0.6 μg/g vs. 12.3 ± 1.9 μg/g, P < 0.01), increased the expression of claudin-5 (0.79 ± 0.08 vs. 0.41 ± 0.06, P < 0.01; 0.97 ± 0.07 vs. 0.41 ± 0.06, P < 0.01), and elevated the ZO-1 protein level (P < 0.05) in brain microvascular segments in a dose-dependent manner in comparison with the corresponding values in the model group. There was no significant difference in occludin expression (P > 0.05). In the hypoxia cell model, NBP pre-treatment improved TJ ultrastructure, decreased intracellular ROS level, and increased the expression of claudin-5 (P < 0.01) and ZO-1 (P < 0.01) in comparison with the corresponding values in the hypoxia group. NBP treatment also elevated the relative expression levels of p-Akt/Akt, p-GSK-3β/GSK-3β, and β-catenin/β-actin in comparison with the corresponding values in the hypoxia group (all P < 0.05). CONCLUSION NBP improves the barrier function of BBB against ischemic injury by upregulating the expression of TJ proteins, possibly by reducing oxidative stress and activating the Akt/GSK-3β/β-catenin signaling pathway.
Collapse
|
20
|
Astaxanthin Ameliorates Ischemic-Hypoxic-Induced Neurotrophin Receptor p75 Upregulation in the Endothelial Cells of Neonatal Mouse Brains. Int J Mol Sci 2019; 20:ijms20246168. [PMID: 31817750 PMCID: PMC6940833 DOI: 10.3390/ijms20246168] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 11/27/2019] [Accepted: 12/03/2019] [Indexed: 12/12/2022] Open
Abstract
Ischemic stroke is a leading cause of human death in present times. Two phases of pathological impact occur during an ischemic stroke, namely, ischemia and reperfusion. Both periods include individual characteristic effects on cell injury and apoptosis. Moreover, these conditions can cause severe cell defects and harm the blood-brain barrier (BBB). Also, the BBB components are the major targets in ischemia-reperfusion injury. The BBB owes its enhanced protective roles to capillary endothelial cells, which maintain BBB permeability. One of the nerve growth factor (NGF) receptors initiating cell signaling, once activated, is the p75 neurotrophin receptor (p75NTR). This receptor is involved in both the survival and apoptosis of neurons. Although many studies have attempted to explain the role of p75NTR in neurons, the mechanisms in endothelial cells remain unclear. Endothelial cells are the first cells to encounter p75NTR stimuli. In this study, we found the upregulated p75NTR expression and reductive expression of tight junction proteins after in vivo and in vitro ischemia-reperfusion injury. Moreover, astaxanthin (AXT), an antioxidant drug, was utilized and was found to reduce p75NTR expression and the number of apoptotic cells. This study verified that p75NTR plays a prominent role in endothelial cell death and provides a novel downstream target for AXT.
Collapse
|
21
|
Acetyl-11-keto-β-boswellic acid (AKBA) Attenuates Oxidative Stress, Inflammation, Complement Activation and Cell Death in Brain Endothelial Cells Following OGD/Reperfusion. Neuromolecular Med 2019; 21:505-516. [DOI: 10.1007/s12017-019-08569-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 09/04/2019] [Indexed: 12/19/2022]
|
22
|
Keratinocyte Growth Factor-2 Is Protective in Oleic Acid-Induced Acute Lung Injury in Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:9406580. [PMID: 31379970 PMCID: PMC6662415 DOI: 10.1155/2019/9406580] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 05/26/2019] [Indexed: 01/04/2023]
Abstract
Objective The aim of this study was to examine the role of keratinocyte growth factor-2 (KGF-2) in oleic acid-induced acute lung injury (ALI) in rats. Methods Forty-five healthy adult male Sprague Dawley rats were divided into 3 groups. Rat ALI model was established by injection of 0.01 mL/kg oleic acid into the tail vein. Rats in the control group were injected with the same amount of normal saline (NS). In the ALI + KGF-2 group, 5 mg/kg of KGF-2 was instilled into the airway of rats 72 hours before the model preparation, and the control group and the ALI model group were instilled with the same amount of NS. The lung permeability index (LPI) and lung wet/dry weight (W / D) ratios were measured 8 hours after the model preparation. The permeability of pulmonary microvascular endothelium was evaluated by Evans blue leakage test. Histopathological changes were observed under light microscope and the ALI pathology score (LIS) was calculated. Ultrastructural changes of lung tissue were observed under electron microscope. The apoptosis was detected by TUNEL assay. The expression of Claudin-5, ZO-1, and VE Cadherin in lung tissue was qualitatively and quantitatively analyzed by immunohistochemistry, Western Blot, and qRT-PCR, respectively. Results The ALI model group had severe lung injury and obvious pathological changes, including alveolar septal thickening and inflammatory cell infiltration. TUNEL assay showed that the apoptosis of ALI group was significantly increased. The LIS score, lung W/D ratio, LPI, and Evans blue leakage were significantly higher than those in the control group; electron microscopy showed that the alveolar-capillary barrier was severely damaged in the ALI group. Compared with the control group, the expression of Claudin-5, ZO-1, and VE cadherin in the lung tissue of the ALI model group was significantly attenuated. After pretreatment with KGF-2, the degree of lung tissue damage was significantly reduced and the pathological changes were significantly improved. TUNEL assay showed that the apoptosis of ALI group was decreased. Lung W/D ratio, LPI, and Evans blue leakage decreased; electron microscopy showed that the alveolar-capillary barrier of ALI group recovered significantly. Compared with the ALI model group, the expression of Claudin-5, ZO-1, and VE cadherin in the lung tissue of the KGF-2 pretreatment group increased. Conclusion The results indicate that KGF-2 may attenuate oleic acid-induced ALI in rats by maintaining the pulmonary microvascular endothelial barrier, which is an effective ALI preventive measure.
Collapse
|
23
|
Crampton AL, Cummins KA, Wood DK. A high-throughput microtissue platform to probe endothelial function in vitro. Integr Biol (Camb) 2019; 10:555-565. [PMID: 30140833 DOI: 10.1039/c8ib00111a] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A critical role of vascular endothelium is as a semi-permeable barrier, dynamically regulating the flux of solutes between blood and the surrounding tissue. Existing platforms that quantify endothelial function in vitro are either significantly throughput limited or overlook physiologically relevant extracellular matrix (ECM) interactions and thus do not recapitulate in vivo function. Leveraging droplet microfluidics, we developed a scalable platform to measure endothelial function in nanoliter-volume, ECM-based microtissues. In this study, we describe our high-throughput method for fabricating endothelial-coated collagen microtissues that incorporate physiologically relevant cell-ECM interactions. We showed that the endothelial cells had characteristic morphology, expressed tight junction proteins, and remodeled the ECM via compaction and deposition of basement membrane. We also measured macromolecular permeability using two optical modalities, and found the cell layers: (1) had permeability values comparable to in vivo measurements and (2) were responsive to physiologically-relevant modulators of endothelial permeability (TNF-α and TGF-β). This is the first demonstration, to the authors' knowledge, of high-throughput assessment (n > 150) of endothelial permeability on natural ECM. Additionally, this technology is compatible with standard cell culture equipment (e.g. multi-well plates) and could be scaled up further to be integrated with automated liquid handling systems and automated imaging platforms. Overall, this platform recapitulates the functions of traditional transwell inserts, but extends application to high-throughput studies and introduces new possibilities for interrogating cell-cell and cell-matrix interactions.
Collapse
Affiliation(s)
- Alexandra L Crampton
- Department of Biomedical Engineering, University of Minnesota, Twin Cities, USA.
| | | | | |
Collapse
|
24
|
Palanisamy K, Karunakaran C, Raman R, Chidambaram S. Optimization of an in vitro bilayer model for studying the functional interplay between human primary retinal pigment epithelial and choroidal endothelial cells isolated from donor eyes. BMC Res Notes 2019; 12:307. [PMID: 31146784 PMCID: PMC6543644 DOI: 10.1186/s13104-019-4333-x] [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] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 05/22/2019] [Indexed: 02/08/2023] Open
Abstract
OBJECTIVE The microenvironment of outer retina is largely regulated by retinal pigment epithelium (RPE) and choroid. Damage to either of these layers lead to the development of age related macular degeneration (AMD). A simplified cell culture model that mimics the RPE/Bruch's membrane (BM) and choroidal layers of the eye is a prerequisite for elucidating the molecular mechanism of disease progression. RESULTS We have isolated primary retinal pigment epithelial cells (hRPE) and human primary choroidal endothelial cells (hCEC) from donor eyes to construct a bilayer of hCEC/hRPE on transwell inserts. Secretion of VEGF in the insert grown bilayer was significantly higher (22 pg/ml) than hCEC monolayer (3 pg/ml). To mimic the disease condition the model was treated with 100 ng/ml of VEGF, which increased the permeability of bilayer for 20 kDa FITC dextran while addition of bevacizumab, a humanized anti-VEGF drug, reversed the effect. To conclude the transwell insert based human primary hCEC/hRPE bilayer model would be an ideal system for studying the disease mechanisms and the crosstalk between RPE and choroid. This model will also be useful in screening small molecules and performing drug permeability kinetics.
Collapse
Affiliation(s)
- Karthikka Palanisamy
- R.S. Mehta Jain Department of Biochemistry and Cell Biology, KBIRVO, Vision Research Foundation, Chennai, India.,School of Chemical and Biotechnology, SASTRA University, Thanjavur, India
| | - Coral Karunakaran
- R.S. Mehta Jain Department of Biochemistry and Cell Biology, KBIRVO, Vision Research Foundation, Chennai, India
| | - Rajiv Raman
- Department of Vitreo-Retinal Services, Medical Research Foundation, Chennai, India
| | - Subbulakshmi Chidambaram
- R.S. Mehta Jain Department of Biochemistry and Cell Biology, KBIRVO, Vision Research Foundation, Chennai, India. .,Department of Biochemistry and Molecular Biology, Pondicherry University, Kalapet, Puducherry, India.
| |
Collapse
|
25
|
Ahmad S, Kindelin A, Khan SA, Ahmed M, Hoda MN, Bhatia K, Ducruet AF. C3a Receptor Inhibition Protects Brain Endothelial Cells Against Oxygen-glucose Deprivation/Reperfusion. Exp Neurobiol 2019; 28:216-228. [PMID: 31138990 PMCID: PMC6526115 DOI: 10.5607/en.2019.28.2.216] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 04/09/2019] [Accepted: 04/10/2019] [Indexed: 02/07/2023] Open
Abstract
The complement cascade is a central component of innate immunity which plays a critical role in brain inflammation. Complement C3a receptor (C3aR) is a key mediator of post-ischemic cerebral injury, and pharmacological antagonism of the C3a receptor is neuroprotective in stroke. Cerebral ischemia injures brain endothelial cells, causing blood brain barrier (BBB) disruption which further exacerbates ischemic neuronal injury. In this study, we used an in vitro model of ischemia (oxygen glucose deprivation; OGD) to investigate the protective effect of a C3aR antagonist (C3aRA, SB290157) on brain endothelial cells (bEnd.3). Following 24 hours of reperfusion, OGD-induced cell death was assessed by TUNEL and Caspase-3 staining. Western blot and immunocytochemistry were utilized to demonstrate that OGD upregulates inflammatory, oxidative stress and antioxidant markers (ICAM-1, Cox-2, Nox-2 and MnSOD) in endothelial cells and that C3aRA treatment significantly attenuate these markers. We also found that C3aRA administration restored the expression level of the tight junction protein occludin in endothelial cells following OGD. Interestingly, OGD/reperfusion injury increased the phosphorylation of ERK1/2 and C3aR inhibition significantly reduced the activation of ERK suggesting that endothelial C3aR may act via ERK signaling. Furthermore, exogenous C3a administration stimulates these same inflammatory mechanisms both with and without OGD, and C3aRA suppresses these C3a-mediated responses, supporting an antagonist role for C3aRA. Based on these results, we conclude that C3aRA administration attenuates inflammation, oxidative stress, ERK activation, and protects brain endothelial cells following experimental brain ischemia.
Collapse
Affiliation(s)
- Saif Ahmad
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Dignity Health, Phoenix, Arizona 85013, USA
| | - Adam Kindelin
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Dignity Health, Phoenix, Arizona 85013, USA
| | - Shah Alam Khan
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Dignity Health, Phoenix, Arizona 85013, USA.,Oman Medical College, Muscat 130, Sultanate of Oman
| | - Maaz Ahmed
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Dignity Health, Phoenix, Arizona 85013, USA
| | - Md Nasrul Hoda
- Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Dignity Health, Phoenix, Arizona 85013, USA
| | - Kanchan Bhatia
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Dignity Health, Phoenix, Arizona 85013, USA.,School of Mathematical and Natural Sciences, Arizona State University, Phoenix, AZ 85004, USA
| | - Andrew F Ducruet
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Dignity Health, Phoenix, Arizona 85013, USA
| |
Collapse
|
26
|
Kunze R, Marti HH. Angioneurins - Key regulators of blood-brain barrier integrity during hypoxic and ischemic brain injury. Prog Neurobiol 2019; 178:101611. [PMID: 30970273 DOI: 10.1016/j.pneurobio.2019.03.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 03/29/2019] [Indexed: 12/14/2022]
Abstract
The loss of blood-brain barrier (BBB) integrity leading to vasogenic edema and brain swelling is a common feature of hypoxic/ischemic brain diseases such as stroke, but is also central to the etiology of other CNS disorders. In the past decades, numerous proteins, belonging to the family of angioneurins, have gained increasing attention as potential therapeutic targets for ischemic stroke, but also other CNS diseases attributed to BBB dysfunction. Angioneurins encompass mediators that affect both neuronal and vascular function. Recently, increasing evidence has been accumulated that certain angioneurins critically determine disease progression and outcome in stroke among others through multifaceted effects on the compromised BBB. Here, we will give a concise overview about the family of angioneurins. We further describe the most important cellular and molecular components that contribute to structural integrity and low permeability of the BBB under steady-state conditions. We then discuss BBB alterations in ischemic stroke, and highlight underlying cellular and molecular mechanisms. For the most prominent angioneurin family members including vascular endothelial growth factors, angiopoietins, platelet-derived growth factors and erythropoietin, we will summarize current scientific literature from experimental studies in animal models, and if available from clinical trials, on the following points: (i) spatiotemporal expression of these factors in the healthy and hypoxic/ischemic CNS, (ii) impact of loss- or gain-of-function during cerebral hypoxia/ischemia for BBB integrity and beyond, and (iii) potential underlying molecular mechanisms. Moreover, we will highlight novel therapeutic strategies based on the activation of endogenous angioneurins that might improve BBB dysfuntion during ischemic stroke.
Collapse
Affiliation(s)
- Reiner Kunze
- Institute of Physiology and Pathophysiology, Heidelberg University, Germany.
| | - Hugo H Marti
- Institute of Physiology and Pathophysiology, Heidelberg University, Germany
| |
Collapse
|
27
|
Aminzadeh A, Salarinejad A. Citicoline protects against lead-induced oxidative injury in neuronal PC12 cells. Biochem Cell Biol 2019; 97:715-721. [PMID: 30925221 DOI: 10.1139/bcb-2018-0218] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Lead is a major environmental pollutant that causes serious adverse effects on biological systems and cells. In this study, we examined the effect of citicoline on lead-induced apoptosis in PC12 cells. The PC12 cells were pre-treated with citicoline and then exposed to lead for 48 h. The effect of citicoline on cell survival was examined by MTT assay. In addition, levels of lipid peroxidation (LPO), total thiol groups, total antioxidant power (TAP), catalase (CAT), superoxide dismutase (SOD), and reduced glutathione (GSH) were evaluated. The levels of Bax, Bcl-2, and caspase-3 were also measured, by Western blot analysis. Citicoline significantly increased the cell viability of PC12 cells exposed to lead. Treatment of PC12 cells with lead increased LPO levels, and citicoline effectively decreased LPO. Levels of total thiol groups and TAP, CAT, SOD, and GSH were significantly increased in citicoline-treated PC12 cells compared with the lead-treated group. Citicoline pretreatment significantly reduced Bax expression, and increased the level of Bcl-2 expression. Citicoline also reduced caspase-3 activation in PC12 cells compared with the lead-treated group. Our findings indicate that citicoline exerts a neuroprotective effect against lead-induced injury in PC12 cells through mitigation of oxidative stress and, at least in part, through suppression of the mitochondria-mediated apoptotic pathway.
Collapse
Affiliation(s)
- Azadeh Aminzadeh
- Department of Pharmacology and Toxicology, School of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran; Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.,Department of Pharmacology and Toxicology, School of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran; Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Ayda Salarinejad
- Department of Pharmacology and Toxicology, School of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran; Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.,Department of Pharmacology and Toxicology, School of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran; Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
28
|
Sun M, Shinoda Y, Fukunaga K. KY-226 Protects Blood-brain Barrier Function Through the Akt/FoxO1 Signaling Pathway in Brain Ischemia. Neuroscience 2018; 399:89-102. [PMID: 30579831 DOI: 10.1016/j.neuroscience.2018.12.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 12/12/2018] [Accepted: 12/16/2018] [Indexed: 01/05/2023]
Abstract
KY-226 is a protein tyrosine phosphatase 1B (PTP1B) inhibitor that protects neurons from cerebral ischemic injury. KY-226 restores Akt (protein kinase B) phosphorylation and extracellular signal-regulated kinase (ERK) reduction in transient middle cerebral artery occlusion (tMCAO) damage. However, the mechanisms underlying the neuroprotective effects of KY-226 are unclear. To address this, the effects of KY-226 on blood-brain barrier (BBB) dysfunction were examined in tMCAO mice. KY-226 (10 mg/kg, i.p.) was administered to ICR mice 30 min after 2 h of tMCAO. To assess Akt or ERK involvement, wortmannin (i.c.v.) or U0126 (i.v.), selective inhibitors of PI3K and ERK, respectively, were administered to mice 30 min before ischemia. BBB integrity was assessed by Evans blue leakage 24 h post-reperfusion. The levels of tight junction (TJ) proteins, ZO-1 and occludin, were measured by western blotting; ZO-1 mRNA level was measured by RT-PCR. Compared to vehicle, KY-226 treatment prevented BBB breakdown and reduction in TJ protein levels. KY-226 treatment restored ZO-1 mRNA levels post-reperfusion. Pre-administration of wortmannin or U0126 blocked the protective effects of KY-226 on ZO-1 protein and mRNA reduction in tMCAO mice. In bEnd.3 cells, lipopolysaccharide treatment reduced mRNA and protein levels of ZO-1, an effect rescued by KY-226 treatment. Further, KY-226 treatment restored phosphorylation of pAkt (T308) and its downstream target forkhead box protein O1 (FoxO1) (S256) in bEnd.3 cells. Collectively, we demonstrate that KY-226 protects BBB integrity by restoration of TJ proteins, an effect partly mediated by Akt/FoxO1 pathway activation. Thus, protection of BBB integrity likely underlies KY-226-induced neuroprotection in tMCAO mice.
Collapse
Affiliation(s)
- Meiling Sun
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai, Japan
| | - Yasuharu Shinoda
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai, Japan
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai, Japan.
| |
Collapse
|
29
|
Leduc-Galindo D, Qvist P, Tóth AE, Fryland T, Nielsen MS, Børglum AD, Christensen JH. The effect of hypoxia on ZEB1 expression in a mimetic system of the blood-brain barrier. Microvasc Res 2018; 122:131-135. [PMID: 30144413 DOI: 10.1016/j.mvr.2018.08.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 05/09/2018] [Accepted: 08/22/2018] [Indexed: 01/29/2023]
Abstract
The blood-brain barrier consists of a tightly sealed monolayer of endothelial cells being vital in maintaining a stable intracerebral microenvironment. The barrier is receptive to leakage upon exposure to environmental factors, like hypoxia, and its disruption has been suggested as a constituent in the pathophysiology of both neurological and psychiatric disorders. The schizophrenia associated ZEB1 gene encodes a transcription factor susceptible to transcriptional control by a hypoxia induced factor, HIF1A, known to be implicated in blood-brain barrier dysfunction. However, whether ZEB1 is also implicated in maintaining blood-brain barrier integrity upon hypoxia is unknown. Here we assessed Hif1a, Zo1 and Zeb1 mRNA expression and ZO1 protein abundancy in a mimetic system of the in vivo blood-brain barrier comprising mouse brain endothelial cells subjected to the norm- and proven hypoxic conditions. Despite that, Hif1a mRNA expression was significantly increased, clearly indicating that the oxygen-deprived environment introduced a hypoxia response in the cells, we found no hypoxia-induced changes in neither ZO1 abundancy nor in the expression of Zo1 and Zeb1 mRNA. However, independent of hypoxia status, we found that Zeb1 and Zo1 mRNA expression is highly correlated. Further studies are warranted that investigate the implication of the ZEB1/ZO1 axis in blood-brain barrier maintenance under different physiological conditions.
Collapse
Affiliation(s)
- Desiree Leduc-Galindo
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; iPSYCH, the Lundbeck Foundation Initiative for Integrative Psychiatric Research, Denmark; iSEQ, Centre for Integrative Sequencing, Aarhus University, Aarhus, Denmark
| | - Per Qvist
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; iPSYCH, the Lundbeck Foundation Initiative for Integrative Psychiatric Research, Denmark; iSEQ, Centre for Integrative Sequencing, Aarhus University, Aarhus, Denmark
| | - Andrea E Tóth
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Tue Fryland
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; iPSYCH, the Lundbeck Foundation Initiative for Integrative Psychiatric Research, Denmark; iSEQ, Centre for Integrative Sequencing, Aarhus University, Aarhus, Denmark
| | | | - Anders D Børglum
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; iPSYCH, the Lundbeck Foundation Initiative for Integrative Psychiatric Research, Denmark; iSEQ, Centre for Integrative Sequencing, Aarhus University, Aarhus, Denmark
| | - Jane H Christensen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; iPSYCH, the Lundbeck Foundation Initiative for Integrative Psychiatric Research, Denmark; iSEQ, Centre for Integrative Sequencing, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
30
|
Luo PL, Wang YJ, Yang YY, Yang JJ. Hypoxia-induced hyperpermeability of rat glomerular endothelial cells involves HIF-2α mediated changes in the expression of occludin and ZO-1. ACTA ACUST UNITED AC 2018; 51:e6201. [PMID: 29791586 PMCID: PMC5972023 DOI: 10.1590/1414-431x20186201] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 03/05/2018] [Indexed: 01/11/2023]
Abstract
This study aimed to investigate the role of hypoxia-inducible factor-2α (HIF-2α) in the expression of tight junction proteins and permeability alterations in rat glomerular endothelial cells (rGENCs) under hypoxia conditions. The expression level of HIF-2α and tight junction proteins (occludin and ZO-1) in rGENCs were examined following 5% oxygen density exposure at different treatment times. HIF-2α lentivirus transfection was used to knockdown HIF-2α expression. Cells were divided into four groups: 1) control group (rGENCs were cultured under normal oxygen conditions), 2) hypoxia group (rGENCs were cultured under hypoxic conditions), 3) negative control group (rGENCs were infected with HIF-2α lentivirus negative control vectors and cultured under hypoxic conditions), and 4) Len group (rGENCs were transfected with HIF-2α lentivirus and cultured under hypoxic conditions). The hypoxia, negative control, and Len groups were kept in a hypoxic chamber (5% O2, 5% CO2, and 90% N2) for 24 h and the total content of occludin and ZO-1, and the permeability of rGENCs were assessed. With increasing hypoxia time, the expression of HIF-2α gradually increased, while the expression of occludin decreased, with a significant difference between groups. ZO-1 expression gradually decreased under hypoxia conditions, but the difference between the 24 and 48 h groups was not significant. The permeability of cells increased following 24-h exposure to hypoxia compared to the control group (P<0.01). The knockdown of HIF-2α expression significantly increased occludin and ZO-1 content compared with hypoxia and negative control groups (P<0.01), while permeability was reduced (P<0.01). Hypoxia increased HIF-2α content, inducing permeability of rGENCs through the reduced expression of occludin and ZO-1.
Collapse
Affiliation(s)
- Peng-Li Luo
- Department of Nephrology, Hospital of Qinghai University, Xining, China
| | - Yan-Jun Wang
- Department of Nephrology, Hospital of Qinghai University, Xining, China
| | - Yan-Yan Yang
- Department of Nephrology, Hospital of Qinghai University, Xining, China
| | - Jia-Jia Yang
- Department of Nephrology, Hospital of Qinghai University, Xining, China
| |
Collapse
|
31
|
Xu M, Yang X, Zeng Q, He H, Lu P, Huang G. BIRC5 is a novel target of peroxisome proliferator‑activated receptor γ in brain microvascular endothelium cells during cerebral ischemia. Mol Med Rep 2017; 16:8882-8890. [PMID: 29039513 PMCID: PMC5779969 DOI: 10.3892/mmr.2017.7750] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 08/16/2017] [Indexed: 11/23/2022] Open
Abstract
Cerebral ischemia is a leading cause of ischemic stroke, which may lead to severe disability and mortality worldwide. There are some key factors concerned in cardioprotection, such as peroxisome proliferator-activated receptor γ (PPARγ), a ligand binding transcription factor involved in various biological functions including atherosclerosis, vascular dysfunction and hypertension, and baculoviral IAP repeat-containing 5 (BIRC5), which may protect human brain endothelial cells from ischemia-induced apoptosis. To determine the potential roles of PPARγ in brain microvascular endothelial (bEnd.3) cells during cerebral ischemia and the relationship between PPARγ and BIRC5, a cerebral ischemia model was established with bEnd.3 cells cells by oxygen-glucose deprivation (OGD) treatment. OGD treatment reduced proliferation and enhanced apoptosis of bEnd.3 cells in a time-dependent manner. PPARγ expression levels were decreased in bEnd.3 cells following OGD treatment. Upregulation of PPARγ expression protected bEnd.3 cells from ischemia injury and also upregulated BIRC5 expression. PPARγ-specific binding sites in the BIRC5 promoter were predicted bioinformatically and verified by luciferase reporter experiments. Results from electrophoretic mobility shift/supershift and chromatin immunoprecipitation assays suggested that BIRC5 may be a novel target of PPARγ transcriptional regulation during ischemic injury. The present results indicated that PPARγ may serve a protective role on bEnd.3 cells and that BIRC5 may be a downstream target of PPARγ regulation during cerebral ischemia.
Collapse
Affiliation(s)
- Mingjing Xu
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - Xianli Yang
- Department of Medical Quality Management, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Qing Zeng
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - He He
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - Pengcheng Lu
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - Guozhi Huang
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| |
Collapse
|
32
|
Trimmel H, Majdan M, Wodak A, Herzer G, Csomor D, Brazinova A. Citicoline in severe traumatic brain injury: indications for improved outcome : A retrospective matched pair analysis from 14 Austrian trauma centers. Wien Klin Wochenschr 2017; 130:37-44. [PMID: 28752349 DOI: 10.1007/s00508-017-1240-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 07/11/2017] [Indexed: 11/27/2022]
Abstract
Goal-oriented management of traumatic brain injury (TBI) can save the lives and/or improve the long-term outcome of millions of affected patients worldwide. Additionally, enhancing quality of life will save enormous socio-economic costs; however, promising TBI treatment strategies with neuroprotective agents, such as citicoline (CDP-choline), lacked evidence or produced contradictory results in clinical trials. During a prehospital TBI project to optimize early TBI care within 14 Austrian trauma centers, data on 778 TBI patients were prospectively collected. As preceding evaluations suggested a beneficial outcome in TBI patients treated at the Wiener Neustadt Hospital (WNH), we aimed to investigate the potential role of citicoline administration, solely applied in WNH, in those patients. In a retrospective subgroup analysis we compared 67 patients from WNH with citicoline administration and 67 matched patients from other Austrian centers without citicoline use. Patients with Glasgow Coma Scale score <13 on site and/or Abbreviated Injury Scale of the region "head" >2 were included. Our analysis revealed significantly reduced rates of intensive care unit (ICU) mortality (5% vs. 24%, p < 0.01), in-hospital mortality (9% vs. 24%, p = 0.035) and 6‑month mortality (13% vs. 28%, p = 0.031), as well as of unfavorable outcome (34% vs. 57%, p = 0.015) and observed vs. expected ratio for mortality (0.42 vs. 0.84) in the WNH (citicoline receivers) group. Despite the limitations of a retrospective subgroup analysis our findings suggest a possible correlation between early and consequent citicoline administration and beneficial outcomes. Therefore, we aim to set up an initiative for a prospective, multicenter randomized controlled trial with citicoline in sTBI (severe TBI) patients.
Collapse
Affiliation(s)
- Helmut Trimmel
- Department of Anesthesiology, Emergency Medicine and Intensive Care and Karl Landsteiner Institute of Emergency Medicine, General Hospital Wiener Neustadt, Wiener Neustadt, Austria.
| | - Marek Majdan
- Department of Public Health, Faculty of Health Sciences and Social Work, Trnava University, Trnava, Slovakia
| | - Andrea Wodak
- Department of Anesthesiology, Emergency Medicine and Intensive Care and Karl Landsteiner Institute of Emergency Medicine, General Hospital Wiener Neustadt, Wiener Neustadt, Austria
| | - Guenther Herzer
- Department of Anesthesiology, Emergency Medicine and Intensive Care and Karl Landsteiner Institute of Emergency Medicine, General Hospital Wiener Neustadt, Wiener Neustadt, Austria
| | - Daniel Csomor
- Department of Anesthesiology, Emergency Medicine and Intensive Care and Karl Landsteiner Institute of Emergency Medicine, General Hospital Wiener Neustadt, Wiener Neustadt, Austria
| | | |
Collapse
|
33
|
miR-Let7A Controls the Cell Death and Tight Junction Density of Brain Endothelial Cells under High Glucose Condition. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:6051874. [PMID: 28680530 PMCID: PMC5478855 DOI: 10.1155/2017/6051874] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 05/07/2017] [Accepted: 05/21/2017] [Indexed: 12/13/2022]
Abstract
Hyperglycemia-induced stress in the brain of patients with diabetes triggers the disruption of blood-brain barrier (BBB), leading to diverse neurological diseases including stroke and dementia. Recently, the role of microRNA becomes an interest in the research for deciphering the mechanism of brain endothelial cell damage under hyperglycemia. Therefore, we investigated whether mircoRNA Let7A (miR-Let7A) controls the damage of brain endothelial (bEnd.3) cells against high glucose condition. Cell viability, cell death marker expressions (p-53, Bax, and cleaved poly ADP-ribose polymerase), the loss of tight junction proteins (ZO-1 and claudin-5), proinflammatory response (interleukin-6, tumor necrosis factor-α), inducible nitric oxide synthase, and nitrite production were confirmed using MTT, reverse transcription-PCR, quantitative-PCR, Western blotting, immunofluorescence, and Griess reagent assay. miR-Let7A overexpression significantly prevented cell death and loss of tight junction proteins and attenuated proinflammatory response and nitrite production in the bEnd.3 cells under high glucose condition. Taken together, we suggest that miR-Let7A may attenuate brain endothelial cell damage by controlling cell death signaling, loss of tight junction proteins, and proinflammatory response against high glucose stress. In the future, the manipulation of miR-Let7A may be a novel solution in controlling BBB disruption which leads to the central nervous system diseases.
Collapse
|
34
|
Liu K, Wang XJ, Li YN, Li B, Qi JS, Zhang J, Wang Y. Tongxinluo Reverses the Hypoxia-suppressed Claudin-9 in Cardiac Microvascular Endothelial Cells. Chin Med J (Engl) 2017; 129:442-7. [PMID: 26879018 PMCID: PMC4800845 DOI: 10.4103/0366-6999.176076] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Background: Claudin-5, claudin-9, and claudin-11 are expressed in endothelial cells to constitute tight junctions, and their deficiency may lead to hyperpermeability, which is the initiating process and pathological basis of cardiovascular disease. Although tongxinluo (TXL) has satisfactory antianginal effects, whether and how it modulates claudin-5, claudin-9, and claudin-11 in hypoxia-stimulated human cardiac microvascular endothelial cells (HCMECs) have not been reported. Methods: In this study, HCMECs were stimulated with CoCl2 to mimic hypoxia and treated with TXL. First, the messenger RNA (mRNA) expression of claudin-5, claudin-9, and claudin-11 was confirmed. Then, the protein content and distribution of claudin-9, as well as cell morphological changes were evaluated after TXL treatment. Furthermore, the distribution and content histone H3K9 acetylation (H3K9ac) in the claudin-9 gene promoter, which guarantees transcriptional activation, were examined to explore the underlying mechanism, by which TXL up-regulates claudin-9 in hypoxia-stimulated HCMECs. Results: We found that hypoxia-suppressed claudin-9 gene expression in HCMECs (F = 7.244; P = 0.011) and the hypoxia-suppressed claudin-9 could be reversed by TXL (F = 61.911; P = 0.000), which was verified by its protein content changes (F = 29.142; P = 0.000). Moreover, high-dose TXL promoted the cytomembrane localization of claudin-9 in hypoxia-stimulated HCMECs, with attenuation of cell injury. Furthermore, high-dose TXL elevated the hypoxia-inhibited H3K9ac in the claudin-9 gene promoter (F = 37.766; P = 0.000), activating claudin-9 transcription. Conclusions: The results manifested that TXL reversed the hypoxia-suppressed claudin-9 by elevating H3K9ac in its gene promoter, playing protective roles in HCMECs.
Collapse
Affiliation(s)
| | | | | | | | - Jin-Sheng Qi
- Department of Biochemistry, Hebei Key Laboratory of Medical Biotechnology, Hebei Medical University, Shijiazhuang, Hebei 050017, China
| | | | | |
Collapse
|
35
|
Li B, Li Y, Liu K, Wang X, Qi J, Wang B, Wang Y. High glucose decreases claudins-5 and -11 in cardiac microvascular endothelial cells: Antagonistic effects of tongxinluo. Endocr Res 2017; 42:15-21. [PMID: 27111519 DOI: 10.3109/07435800.2016.1163723] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
UNLABELLED Purpose/aim of the study: Claudins-5, -9, and -11 are tight-junction proteins that are mainly expressed in endothelial cells. Their deficiency may lead to cell barrier dysfunction, which is considered as the initiating process and pathological basis of cardiovascular disease in diabetes. We investigated whether high glucose (HG) affects claudins-5, -9, and -11 in human cardiac microvascular endothelial cells (HCMECs), and examined the effects of the traditional Chinese medication tongxinluo (TXL) on these tight junction proteins. MATERIALS AND METHODS HCMECs were exposed to HG with and without TXL treatment, and then mRNA and protein levels of claudins-5, -9, and -11 were examined. The distribution of claudins-5 and -11 was also investigated. Histone H3K9 acetylation (H3K9ac) in claudin-5 and claudin-11 gene promoters, which functions in transactivation, was measured. RESULTS We found that HG suppressed claudins-5 and -11 gene expression in HCMECs, and TXL reversed the HG-mediated inhibition of claudins-5 and -11 mRNA and protein expressions. Treatment with high-dose of TXL promoted cell membrane localization of claudins-5 and -11 in HG-stimulated HCMECs. Furthermore, high-dose of TXL blocked the inhibition of H3K9ac in claudin-5 and claudin-11 gene promoters caused by exposure to HG, thus activating gene transcription. CONCLUSIONS Our results show that HG suppressed claudins-5 and -11 in HCMECs, and TXL could reverse the HG-induced suppression of claudins-5 and -11 by increasing H3K9ac in their respective gene promoters.
Collapse
Affiliation(s)
- Bin Li
- a Department of Biochemistry , Hebei Key Laboratory of Medical Biotechnology , Shijiazhuang , PR China
| | - Yanning Li
- b Department of Molecular Biology , Hebei Key Lab of Laboratory Animal, Hebei Medical University , Shijiazhuang , PR China
| | - Kun Liu
- a Department of Biochemistry , Hebei Key Laboratory of Medical Biotechnology , Shijiazhuang , PR China
| | - Xiujuan Wang
- a Department of Biochemistry , Hebei Key Laboratory of Medical Biotechnology , Shijiazhuang , PR China
| | - Jinsheng Qi
- a Department of Biochemistry , Hebei Key Laboratory of Medical Biotechnology , Shijiazhuang , PR China
| | - Boya Wang
- a Department of Biochemistry , Hebei Key Laboratory of Medical Biotechnology , Shijiazhuang , PR China
| | - Yu Wang
- b Department of Molecular Biology , Hebei Key Lab of Laboratory Animal, Hebei Medical University , Shijiazhuang , PR China
| |
Collapse
|
36
|
Hao L, Guo X, Zou C, Zhou H, Tian H, Zhang Y, Song C, Liu L. Hyperbaric oxygen preconditioning ameliorates blood-brain barrier damage induced by hypoxia through modulation of tight junction proteins in an in vitro model. Croat Med J 2016; 57:51-7. [PMID: 26935614 PMCID: PMC4800327 DOI: 10.3325/cmj.2016.57.51] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Aim To explore the effects of hyperbaric oxygen preconditioning (HBOP) on the permeability of blood-brain barrier (BBB) and expression of tight junction proteins under hypoxic conditions in vitro. Methods A BBB in vitro model was constructed using the hCMEC/D3 cell line and used when its trans-endothelial electrical resistance (TEER) reached 80-120 Ω · cm2 (tested by Millicell-Electrical Resistance System). The cells were randomly divided into the control group cultured under normal conditions, the group cultured under hypoxic conditions (2%O2) for 24 h (hypoxia group), and the group first subjected to HBOP for 2 h and then to hypoxia (HBOP group). Occludin and ZO-1 expression were analyzed by immunofluorescence assay. Results Normal hCMEC/D3 was spindle-shaped and tightly integrated. TEER was significantly reduced in the hypoxia (P = 0.001) and HBOP group (P = 0.014) compared to control group, with a greater decrease in the hypoxia group. Occludin membranous expression was significantly decreased in the hypoxia group (P = 0.001) compared to the control group, but there was no change in the HBOP group. ZO-1 membranous expression was significantly decreased (P = 0.002) and cytoplasmic expression was significantly increased (P = 0.001) in the hypoxia group compared to the control group, although overall expression levels did not change. In the HBOP group, there was no significant change in ZO-1 expression compared to the control group. Conclusion Hyperbaric oxygen preconditioning protected the integrity of BBB in an in vitro model through modulation of occludin and ZO-1 expression under hypoxic conditions.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Lei Liu
- Lei Liu, No. 29 Jianxin East Road, Jiangbei District, Chongqing, 400020, China,
| |
Collapse
|
37
|
Ye LB, Yu XC, Xia QH, Yang Y, Chen DQ, Wu F, Wei XJ, Zhang X, Zheng BB, Fu XB, Xu HZ, Li XK, Xiao J, Zhang HY. Regulation of Caveolin-1 and Junction Proteins by bFGF Contributes to the Integrity of Blood-Spinal Cord Barrier and Functional Recovery. Neurotherapeutics 2016; 13:844-858. [PMID: 27170156 PMCID: PMC5147725 DOI: 10.1007/s13311-016-0437-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The blood-spinal cord barrier (BSCB) plays important roles in the recovery of spinal cord injury (SCI), and caveolin-1 is essential for the integrity and permeability of barriers. Basic fibroblast growth factor (bFGF) is an important neuroprotective protein and contributes to the survival of neuronal cells. This study was designed to investigate whether bFGF is beneficial for the maintenance of junction proteins and the integrity of the BSCB to identify the relations with caveolin-1 regulation. We examined the integrity of the BSCB with Evans blue dye and fluorescein isothiocyanate-dextran extravasation, measured the junction proteins and matrix metalloproteinases, and evaluated the locomotor function recovery. Our data indicated that bFGF treatment improved the recovery of BSCB and functional locomotion in contusive SCI model rats, reduced the expression and activation of matrix metalloproteinase-9, and increased the expressions of caveolin-1 and junction proteins, including occludin, claudin-5, p120-catenin, and β-catenin. In the brain, in microvascular endothelial cells, bFGF treatment increased the levels of junction proteins, caveolin-1 small interfering RNA abolished the protective effect of bFGF under oxygen-glucose deprivation conditions, and the expression of fibroblast growth factor receptor 1 and co-localization with caveolin-1 decreased significantly, which could not be reversed by bFGF treatment. These findings provide a novel mechanism underlying the beneficial effects of bFGF on the BSCB and recovery of SCI, especially the regulation of caveolin-1.
Collapse
Affiliation(s)
- Li-Bing Ye
- School of Pharmaceutical Sciences, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Xi-Chong Yu
- School of Pharmaceutical Sciences, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Qing-Hai Xia
- School of Pharmaceutical Sciences, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Ying Yang
- School of Pharmaceutical Sciences, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Da-Qing Chen
- Emergency Department, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Fenzan Wu
- Department of Neurosurgery, Cixi People's Hospital, Ningbo, Zhejiang, 315300, China
| | - Xiao-Jie Wei
- Department of Neurosurgery, Cixi People's Hospital, Ningbo, Zhejiang, 315300, China
| | - Xie Zhang
- Ningbo Medical Treatment Center, Li Huili Hospital, Ningbo, Zhejiang, 315000, China
| | - Bin-Bin Zheng
- Department of Orthopaedics, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Xiao-Bing Fu
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing, 100048, China
| | - Hua-Zi Xu
- Department of Orthopaedics, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Xiao-Kun Li
- School of Pharmaceutical Sciences, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Jian Xiao
- School of Pharmaceutical Sciences, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| | - Hong-Yu Zhang
- School of Pharmaceutical Sciences, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| |
Collapse
|
38
|
A New Explanation of Inflammation in Rheumatoid Arthritis Patients With Respect to Claudin-5, Matrix Metalloproteinase-9, and Neuroserpin. Arch Rheumatol 2016; 31:299-305. [PMID: 30375560 DOI: 10.5606/archrheumatol.2016.5974] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 04/03/2016] [Indexed: 11/21/2022] Open
Abstract
Objectives This study aims to investigate the relationship between neuroserpin (NSP) and claudin-5, as well as matrix metalloproteinase-9 (MMP-9), with respect to clinical activity of disease in patients with rheumatoid arthritis. Patients and methods The study included a total of 75 patients (18 males, 57 females; mean age 48.12±11.23 years; range 20 to 60 years) who were admitted to the rheumatology outpatient facility at the Medical Faculty Hospital, Sakarya University, in October 2014. Patients were divided into four groups based on their Disease Activity Score 28 (DAS28) scores as remission group (n=16, DAS28 <2.6), low disease activity group (n=16, DAS28 between 2.6-3.2), moderate disease activity group (n=28, DAS28 between 3.2-5.1), and high disease activity group (n=15, DAS28 >5.1). Ten healthy subjects (HS) served as controls. Results Claudin-5, MMP-9, and NSP levels were significantly different in rheumatoid arthritis patients compared to HS (p=0.035, 0.026, and 0.014, respectively). Additionally, there were no differences between claudin-5 levels and disease activity among all RA groups. However, compared to HS, patient groups showed a significant difference (p=0.035) in terms of claudin-5 levels. Serum levels of MMP-9 were significantly different in moderate disease activity group compared to HS (p=0.013). Levels of NSP were significantly different in moderate disease activity and high disease activity groups compared to HS (p=0.008 and 0.031, respectively). Conclusion Our study demonstrated the differential associations of endothelial function/dysfunction biomarkers and disease activity in rheumatoid arthritis. How and why this impairment occurs is not fully understood and more data regarding NSP, MMP, and claudin expression in plasma are warranted.
Collapse
|
39
|
Pan Q, He C, Liu H, Liao X, Dai B, Chen Y, Yang Y, Zhao B, Bihl J, Ma X. Microvascular endothelial cells-derived microvesicles imply in ischemic stroke by modulating astrocyte and blood brain barrier function and cerebral blood flow. Mol Brain 2016; 9:63. [PMID: 27267759 PMCID: PMC4897950 DOI: 10.1186/s13041-016-0243-1] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 05/25/2016] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Endothelial cell (EC) released microvesicles (EMVs) can affect various target cells by transferring carried genetic information. Astrocytes are the main components of the blood brain barrier (BBB) structure in the brain and participate in regulating BBB integrity and blood flow. The interactions between ECs and astrocytes are essential for BBB integrity in homeostasis and pathological conditions. Here, we studied the effects of human brain microvascular ECs released EMVs on astrocyte functions. Additionally, we investigated the effects of EMVs treated astrocytes on regulating BBB function and cerebral ischemic damage. RESULTS EMVs prepared from ECs cultured in normal condition (n-EMVs) or oxygen and glucose deprivation (OGD-EMVs) condition had diverse effects on astrocytes. The n-EMVs promoted, while the OGD-EMVs inhibited the proliferation of astrocytes via regulating PI3K/Akt pathway. Glial fibrillary acidic protein (GFAP) expression (marker of astrocyte activation) was up-regulated by n-EMVs, while down-regulated by OGD-EMVs. Meanwhile, n-EMVs inhibited but OGD-EMVs promoted the apoptosis of astrocytes accompanied by up/down-regulating the expression of Caspase-9 and Bcl-2. In the BBB model of ECs-astrocytes co-culture, the n-EMVs, conversely to OGD-EMVs, decreased the permeability of BBB accompanied with up-regulation of zonula occudens-1(ZO-1) and Claudin-5. In a transient cerebral ischemia mouse model, n-EMVs ameliorated, while OGD-EMVs aggravated, BBB disruption, local cerebral blood flow (CBF) reduction, infarct volume and neurological deficit score. CONCLUSIONS Our data suggest that EMVs diversely modulate astrocyte functions, BBB integrity and CBF, and could serve as a novel therapeutic target for ischemic stroke.
Collapse
Affiliation(s)
- Qunwen Pan
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Caixia He
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China.,Guangdong Medical University, Zhanjiang, 524001, China
| | - Hua Liu
- College of Health Science, Wuhan Sports University, Wuhan, Hubei, 430000, China
| | - Xiaorong Liao
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Bingyan Dai
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Yanfang Chen
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China.,Department of Pharmacology and Toxicology, Wright State University, Dayton, OH, 45435, USA.,Department of Neurology, Boonshoft School of Medicine, Wright State University, Dayton, OH, 45435, USA
| | - Yi Yang
- College of Health Science, Wuhan Sports University, Wuhan, Hubei, 430000, China
| | - Bin Zhao
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Ji Bihl
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China. .,Department of Pharmacology and Toxicology, Wright State University, Dayton, OH, 45435, USA.
| | - Xiaotang Ma
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China.
| |
Collapse
|
40
|
He X, Li H, Chen Y, Chen A, Shan K, Chen J, Zhao H, Zhang X, Cai T. The Effects of IL-22 on the Inflammatory Mediator Production, Proliferation, and Barrier Function of HUVECs. Inflammation 2016; 39:1099-107. [PMID: 27059499 DOI: 10.1007/s10753-016-0341-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The aim of this study was to investigate the effects of interleukin (IL)-22 on proliferation function and inflammatory mediator production and barrier function of human umbilical vein endothelial cells (HUVECs). The expression of mRNA was detected by RT-PCR. The proliferation ability of cells was evaluated using a cell counting kit assay. Real-time quantitative PCR and Western blot were used to detect the expression of inflammatory mediators. The endothelial barrier permeability was assessed by measuring permeability to FITC-labeled dextran. The distribution of occludin was detected by immunofluorescence. IL-22R1 mRNA expression was noted in HUVECs. IL-22 could enhance the proliferation ability of HUVECs and suppress lipopolysaccharide (LPS)-induced proliferation inhibition in these cells. IL-22 also enhanced the production of CCL2 and CCL20 by HUVECs. Besides, IL-22 could improve barrier function and decrease LPS-induced increased cellular permeability and inhibit the LPS-induced destruction of occludin in HUVECs. IL-22 may play a protective role in the development of vasculitis.
Collapse
Affiliation(s)
- Xian He
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Hui Li
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Ying Chen
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Aijun Chen
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Kui Shan
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Jin Chen
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Hengguang Zhao
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Xiaojiao Zhang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Tao Cai
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.
| |
Collapse
|
41
|
Tongxinluo (TXL), a Traditional Chinese Medicinal Compound, Improves Endothelial Function After Chronic Hypoxia Both In Vivo and In Vitro. J Cardiovasc Pharmacol 2016; 65:579-86. [PMID: 26065642 PMCID: PMC4461393 DOI: 10.1097/fjc.0000000000000226] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Vascular injury after chronic hypoxia leads to endothelial injury and structural damage to tight junctions (TJs), thereby resulting in a variety of cardiovascular diseases. Thus, attenuating hypoxia-induced damage has great significance for the prevention and treatment of cardiovascular disease. The aim of this study was to investigate whether the endothelial protection conferred by tongxinluo (TXL), a traditional Chinese medicinal compound, is related to its regulation of TJ protein expression. In vivo, we found that TXL could promote hypoxia-induced angiogenesis in lung and liver tissue. In vitro, we found that CoCl2 treatment significantly reduced the expression of the TJ proteins occludin, claudin-1, VE-cadherin, and beta-catenin in cultured human cardiac microvascular endothelial cells. TXL pretreatment abrogated the CoCl2-induced downregulation of these TJ proteins. Conversely, overexpression of Krüppel-like factor 4 (KLF4) inhibited the expression of TJ proteins in human cardiac microvascular endothelial cells, an effect that was reversed by TXL pretreatment. Further experiments showed that TXL could promote endothelial cell proliferation by increasing KLF4 phosphorylation, thereby reversing the effect of KLF4 on the expression of TJ proteins. These findings provide a new molecular mechanism for the TXL-induced increase in TJ protein expression.
Collapse
|
42
|
Hu J, Ni S, Cao Y, Zhang T, Wu T, Yin X, Lang Y, Lu H. The Angiogenic Effect of microRNA-21 Targeting TIMP3 through the Regulation of MMP2 and MMP9. PLoS One 2016; 11:e0149537. [PMID: 26872030 PMCID: PMC4752282 DOI: 10.1371/journal.pone.0149537] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 02/02/2016] [Indexed: 11/18/2022] Open
Abstract
microRNAs are a novel set of small, non-protein-coding nucleotide RNAs that negatively regulate the expression of target mRNAs. miRNA-21 is a microRNA that is highly enriched in endothelial cells. miRNA-21 has been shown to be a potential pro-angiogenic factor in some biological systems. Our previous study showed that the expression of miRNA-21 was up-regulated after spinal cord injury. However, the effect of miRNA-21 on angiogenesis in the spinal cord was unclear. In this study, to understand the role of miRNA-21 on injured endothelial cells exclusively, an oxygen and glucose deprivation model of endothelial cells was constructed, and the up-regulation of miRNA-21 was discovered in this model. An increased level of miRNA-21 by mimics promoted the survival, migration and tube formation of endothelial cells, which simultaneously inhibited tissue inhibitor of metalloproteinase-3 (TIMP3) expression and promoted matrix metalloproteinase-2 (MMP2) and matrix metalloproteinase-9 (MMP9) expression and secretion. A decreased level of miRNA-21 by antagomir exerted an opposite effect. As is well known, survival, migration and tube formation of endothelial cells are necessary prerequisites for angiogenesis after injury. TIMP3 was validated as a direct target of miRNA-21 by dual-luciferase reporter assay. Silencing with small interfering RNA against TIMP3 promoted tube formation and increased MMP2 and MMP9 expression at the protein level. In vivo, we found that decreased levels of miRNA-21 inhibited angiogenesis after spinal cord injury in rats using synchrotron radiation micro-computed tomography. In summary, these findings suggest that miRNA-21 has a protective effect on angiogenesis by reducing cell death and promoting cell survival, migration and tube formation via partially targeting the TIMP3 by potentially regulating MMP2 and MMP9. TIMP3 is a functional target gene. Identifying the role of miRNA-21 in the protection of angiogenesis might offer a novel therapeutic target for secondary spinal cord injury, in which angiogenesis is indispensable.
Collapse
Affiliation(s)
- Jianzhong Hu
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, 410008, PR China
| | - Shuangfei Ni
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, 410008, PR China
| | - Yong Cao
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, 410008, PR China
| | - Tao Zhang
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, 410008, PR China
| | - Tianding Wu
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, 410008, PR China
| | - Xianzhen Yin
- Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, PR China
| | - Ye Lang
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, 410008, PR China
| | - Hongbin Lu
- Department of Sports Medicine, Research Centre of Sports Medicine, Xiangya Hospital, Central South University, Changsha, 410008, PR China
- * E-mail:
| |
Collapse
|
43
|
Dong W, Xiao S, Cheng M, Ye X, Zheng G. Minocycline induces protective autophagy in vascular endothelial cells exposed to an in vitro model of ischemia/reperfusion-induced injury. Biomed Rep 2015; 4:173-177. [PMID: 26893833 PMCID: PMC4734200 DOI: 10.3892/br.2015.554] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 10/23/2015] [Indexed: 11/17/2022] Open
Abstract
Minocycline has been reported to exhibit advantageous effects on ischemic stroke; however, the precise mechanism of minocycline remains to be established. In the present study, human umbilical vein endothelial cells (HUVECs) were subjected to in vitro simulated ischemia/reperfusion conditions to determine the potential effect of minocycline-induced autophagy on HUVEC damage under oxygen-glucose deprivation/reperfusion (OGD/R). The study demonstrated that minocycline enhanced autophagy in a dose-dependent manner in HUVECs exposed to OGD/R, and only low-dose minocycline protected HUVECs from OGD/R-induced damage. Subsequently, 3-methyladenine (3-MA) was added into the culture media and the protective effect of minocycline was abolished. At the same time, it has been observed that simultaneous treatment with 3-MA also inhibited the autophagy activity induced by minocycline. This finding could suggest that autophagy induced by minocycline serves as one of the potential protective mechanism underlying the beneficial effects of minocycline on ischemic injury.
Collapse
Affiliation(s)
- Wenbin Dong
- Institute of Materia Medica, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, P.R. China
| | - Shigeng Xiao
- Institute of Materia Medica, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, P.R. China
| | - Min Cheng
- Institute of Materia Medica, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, P.R. China
| | - Xiaodi Ye
- Institute of Materia Medica, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, P.R. China
| | - Gaoli Zheng
- Institute of Materia Medica, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, P.R. China
| |
Collapse
|
44
|
Wang M, Chen M, Ding Y, Zhu Z, Zhang Y, Wei P, Wang J, Qiao Y, Li L, Li Y, Wen A. Pretreatment with β-Boswellic Acid Improves Blood Stasis Induced Endothelial Dysfunction: Role of eNOS Activation. Sci Rep 2015; 5:15357. [PMID: 26482008 PMCID: PMC4611516 DOI: 10.1038/srep15357] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 09/22/2015] [Indexed: 11/23/2022] Open
Abstract
Vascular endothelial cells play an important role in modulating anti-thrombus and maintaining the natural function of vascular by secreting many active substances. β-boswellic acid (β-BA) is an active triterpenoid compound from the extract of boswellia serrate. In this study, it is demonstrated that β-BA ameliorates plasma coagulation parameters, protects endothelium from blood stasis induced injury and prevents blood stasis induced impairment of endothelium-dependent vasodilatation. Moreover, it is found that β-BA significantly increases nitric oxide (NO) and cyclic guanosine 3’, 5’-monophosphate (cGMP) levels in carotid aortas of blood stasis rats. To stimulate blood stasis-like conditions in vitro, human umbilical vein endothelial cells (HUVECs) were exposed to transient oxygen and glucose deprivation (OGD). Treatment of β-BA significantly increased intracellular NO level. Western blot and immunofluorescence as well as immunohistochemistry reveal that β-BA increases phosphorylation of enzyme nitric oxide synthase (eNOS) at Ser1177. In addition, β-BA mediated endothelium-dependent vasodilatation can be markedly blocked by eNOS inhibitor L-NAME in blood stasis rats. In OGD treated HUEVCs, the protective effect of β-BA is attenuated by knockdown of eNOS. In conclusion, the above findings provide convincing evidence for the protective effects of β-BA on blood stasis induced endothelial dysfunction by eNOS signaling pathway.
Collapse
Affiliation(s)
- Mingming Wang
- Department of pharmacy, Xijing Hospital, Fourth Military Medical University, Shaanxi, Xi'an 710032, China.,Shaanxi University of Chinese Medicine, Shaanxi, Xian-yang 712046, China
| | - Minchun Chen
- Department of pharmacy, Xijing Hospital, Fourth Military Medical University, Shaanxi, Xi'an 710032, China
| | - Yi Ding
- Department of pharmacy, Xijing Hospital, Fourth Military Medical University, Shaanxi, Xi'an 710032, China
| | - Zhihui Zhu
- Department of pharmacy, Xijing Hospital, Fourth Military Medical University, Shaanxi, Xi'an 710032, China
| | - Yikai Zhang
- Department of pharmacy, Xijing Hospital, Fourth Military Medical University, Shaanxi, Xi'an 710032, China
| | - Peifeng Wei
- Shaanxi University of Chinese Medicine, Shaanxi, Xian-yang 712046, China
| | - Jingwen Wang
- Department of pharmacy, Xijing Hospital, Fourth Military Medical University, Shaanxi, Xi'an 710032, China
| | - Yi Qiao
- Department of pharmacy, Xijing Hospital, Fourth Military Medical University, Shaanxi, Xi'an 710032, China
| | - Liang Li
- Department of pharmacy, Xijing Hospital, Fourth Military Medical University, Shaanxi, Xi'an 710032, China
| | - Yuwen Li
- Department of pharmacy, Xijing Hospital, Fourth Military Medical University, Shaanxi, Xi'an 710032, China
| | - Aidong Wen
- Department of pharmacy, Xijing Hospital, Fourth Military Medical University, Shaanxi, Xi'an 710032, China
| |
Collapse
|
45
|
Martynov MY, Gusev EI. Current knowledge on the neuroprotective and neuroregenerative properties of citicoline in acute ischemic stroke. J Exp Pharmacol 2015; 7:17-28. [PMID: 27186142 PMCID: PMC4863531 DOI: 10.2147/jep.s63544] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Ischemic stroke is one of the leading causes of long-lasting disability and death. Two main strategies have been proposed for the treatment of ischemic stroke: restoration of blood flow by thrombolysis or mechanical thrombus extraction during the first few hours of ischemic stroke, which is one of the most effective treatments and leads to a better functional and clinical outcome. The other direction of treatment, which is potentially applicable to most of the patients with ischemic stroke, is neuroprotection. Initially, neuroprotection was mainly targeted at protecting gray matter, but during the past few years there has been a transition from a neuron-oriented approach toward salvaging the whole neurovascular unit using multimodal drugs. Citicoline is a multimodal drug that exhibits neuroprotective and neuroregenerative effects in a variety of experimental and clinical disorders of the central nervous system, including acute and chronic cerebral ischemia, intracerebral hemorrhage, and global cerebral hypoxia. Citicoline has a prolonged therapeutic window and is active at various temporal and biochemical stages of the ischemic cascade. In acute ischemic stroke, citicoline provides neuroprotection by attenuating glutamate exitotoxicity, oxidative stress, apoptosis, and blood–brain barrier dysfunction. In the subacute and chronic phases of ischemic stroke, citicoline exhibits neuroregenerative effects and activates neurogenesis, synaptogenesis, and angiogenesis and enhances neurotransmitter metabolism. Acute and long-term treatment with citicoline is safe and in most clinical studies is effective and improves functional outcome.
Collapse
Affiliation(s)
- Mikhail Yu Martynov
- Department of Neurology, Neurosurgery and Medical Genetics, Russian National Research Medical University, Moscow, Russia
| | - Eugeny I Gusev
- Department of Neurology, Neurosurgery and Medical Genetics, Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
46
|
Sun L, Liu S, Bao L, Li Y, Feng J, Liu Z. Claudin multigene family in channel catfish and their expression profiles in response to bacterial infection and hypoxia as revealed by meta-analysis of RNA-Seq datasets. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2015; 13:60-9. [PMID: 25681604 DOI: 10.1016/j.cbd.2015.01.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2014] [Revised: 01/14/2015] [Accepted: 01/14/2015] [Indexed: 11/19/2022]
Abstract
Claudins are one of the major groups of transmembrane proteins that play crucial roles in tight junctions. In addition to their function in the regulation of paracellular permeability, claudins are also involved in a number of biological processes related to pathogen infection, embryonic development, organ development and hypoxia response. Despite its importance, analyses of claudin genes in channel catfish have not been systematically performed. In this study, a total of 52 claudin genes were identified and characterized in channel catfish. Phylogenetic analyses were conducted to determine their identities and identify a number of lineage-specific claudin gene duplications in channel catfish. Expression profiles of catfish claudin genes in response to enteric septicemia of catfish (ESC) disease and hypoxia stress were determined by analyzing existing RNA-Seq datasets. Claudin genes were significantly down-regulated in the intestine at 3h post-infection, indicating that pathogens may disrupt the mucosal barrier by suppressing the expression of claudin genes. A total of six claudin genes were significantly regulated in the gill after hypoxia stress. Among them, the expressions of cldn-11b and cldn-10d were dramatically altered when comparing hypoxia tolerant fish with intolerant fish, though their specific roles involved in response to hypoxia stress remained unknown.
Collapse
Affiliation(s)
- Luyang Sun
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, Program of Cell and Molecular Biosciences, Auburn University, Auburn, AL 36849, USA
| | - Shikai Liu
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, Program of Cell and Molecular Biosciences, Auburn University, Auburn, AL 36849, USA
| | - Lisui Bao
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, Program of Cell and Molecular Biosciences, Auburn University, Auburn, AL 36849, USA
| | - Yun Li
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, Program of Cell and Molecular Biosciences, Auburn University, Auburn, AL 36849, USA
| | - Jianbin Feng
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, Program of Cell and Molecular Biosciences, Auburn University, Auburn, AL 36849, USA
| | - Zhanjiang Liu
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, Program of Cell and Molecular Biosciences, Auburn University, Auburn, AL 36849, USA.
| |
Collapse
|