51
|
Heo SH, Song J, Kim BJ, Kim H, Chang DI. Rationale and design to assess the efficacy and safety of HT047 in patients with acute ischemic stroke: A multicenter, randomized, double-blind, placebo-controlled, parallel-group, phase II trial. Medicine (Baltimore) 2019; 98:e17655. [PMID: 31651889 PMCID: PMC6824685 DOI: 10.1097/md.0000000000017655] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Though several neuroprotective agents have been evaluated as potential treatments for acute ischemic stroke, none have demonstrated a definitive treatment efficacy, which remains elusive. HT047 is an herbal extract of Scutellaria baicalensis and Pueraria lobata, both of which have been widely used to treat ischemic stroke in traditional Korean medicine. The aims of this trial are to investigate whether HT047 can improve neurologic status, particularly motor function, in acute ischemic stroke patients, and to determine the safety of HT047. METHODS A multicenter, double-blind, randomized, placebo-controlled, 3-arm parallel group, phase II trial will be conducted in patients who have had an acute ischemic stroke within the past 14 days. The participating patients must have a Fugl-Meyer assessment (FMA) motor score ≤55, with arm or leg weakness, and Korean version of the National Institutes of Health Stroke scale (K-NIHSS) score of ≥4 and ≤15. Seventy-eight participants will be randomized in a 1:1:1 ratio and given high-dose HT047 (750 mg 3 times a day), low-dose HT047 (500 mg 3 times a day), or a placebo for 12 weeks. The primary endpoint is the change in FMA motor score between baseline and week 12. Secondary endpoints are as follows: the change in FMA motor score at weeks 4 and 8 from baseline; the change in FMA motor score at weeks 4, 8, and 12 from baseline according to the timing of treatment initiation (either within 1 week, or 1-2 weeks), or according to the presence of prognostic risk factors (hypertension, diabetes, dyslipidemia, etc); the change in K-NIHSS and Korean versions of the modified Rankin scale (K-mRS) and the modified Barthel index at weeks 4 and 12 from baseline; and the proportion of subjects at week 12 with a K-NIHSS score of 0 to 2, or with K-mRS scores of 0, ≤1, and ≤2. DISCUSSION This study is a 1st-in-human trial of HT047 to explore the efficacy and safety in acute ischemic stroke patients. The results will provide the appropriate dosage and evidence of therapeutic benefit of HT047 for stroke recovery. TRIAL REGISTRATION ClinicalTrials.gov (NCT02828540) Registered July 11, 2016.
Collapse
Affiliation(s)
- Sung Hyuk Heo
- Department of Neurology, Kyung Hee University Hospital
| | - Jungbin Song
- Department of Herbal Pharmacology, Kyung Hee University College of Korean Medicine, Seoul, Republic of Korea
| | - Bum Joon Kim
- Department of Neurology, Kyung Hee University Hospital
| | - Hocheol Kim
- Department of Herbal Pharmacology, Kyung Hee University College of Korean Medicine, Seoul, Republic of Korea
| | - Dae-Il Chang
- Department of Neurology, Kyung Hee University Hospital
| |
Collapse
|
52
|
Sun X, Jung JH, Arvola O, Santoso MR, Giffard RG, Yang PC, Stary CM. Stem Cell-Derived Exosomes Protect Astrocyte Cultures From in vitro Ischemia and Decrease Injury as Post-stroke Intravenous Therapy. Front Cell Neurosci 2019; 13:394. [PMID: 31551712 PMCID: PMC6733914 DOI: 10.3389/fncel.2019.00394] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 08/13/2019] [Indexed: 01/13/2023] Open
Abstract
In the present study, we assessed efficacy of exosomes harvested from human and mouse stem cell cultures in protection of mouse primary astrocyte and neuronal cell cultures following in vitro ischemia, and against ischemic stroke in vivo. Cell media was collected from primary mouse neural stem cell (NSC) cultures or from human induced pluripotent stem cell-derived cardiomyocyte (iCM) cultures. Exosomes were extracted and purified by polyethylene glycol complexing and centrifugation, and exosome size and concentration were determined with a NanoSiteTM particle analyzer. Exosomes were applied to primary mouse cortical astrocyte or neuronal cultures prior to, and/or during, combined oxygen-glucose deprivation (OGD) injury. Cell death was assessed via lactate dehydrogenase (LHD) and propidium iodide staining 24 h after injury. NSC-derived exosomes afforded marked protection to astrocytes following OGD. A more modest (but significant) level of protection was observed with human iCM-derived exosomes applied to astrocytes, and with NSC-derived exosomes applied to primary neuronal cultures. In subsequent experiments, NSC-derived exosomes were injected intravenously into adult male mice 2 h after transient (1 h) middle cerebral artery occlusion (MCAO). Gross motor function was assessed 1 day after reperfusion and infarct volume was assessed 4 days after reperfusion. Mice treated post-stroke with intravenous NSC-derived exosomes exhibited significantly reduced infarct volumes. Together, these results suggest that exosomes isolated from mouse NSCs provide neuroprotection against experimental stroke possibly via preservation of astrocyte function. Intravenous NSC-derived exosome treatment may therefore provide a novel clinical adjuvant for stroke in the immediate post-injury period.
Collapse
Affiliation(s)
- Xiaoyun Sun
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Ji-Hye Jung
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States.,Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States
| | - Oiva Arvola
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Michelle R Santoso
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Rona G Giffard
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Phillip C Yang
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States.,Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States
| | - Creed M Stary
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
53
|
Role of exosomes induced by remote ischemic preconditioning in neuroprotection against cerebral ischemia. Neuroreport 2019; 30:834-841. [DOI: 10.1097/wnr.0000000000001280] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
54
|
Durán-Laforet V, Fernández-López D, García-Culebras A, González-Hijón J, Moraga A, Palma-Tortosa S, García-Yébenes I, Vega-Pérez A, Lizasoain I, Moro MÁ. Delayed Effects of Acute Reperfusion on Vascular Remodeling and Late-Phase Functional Recovery After Stroke. Front Neurosci 2019; 13:767. [PMID: 31396042 PMCID: PMC6664024 DOI: 10.3389/fnins.2019.00767] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Accepted: 07/09/2019] [Indexed: 11/24/2022] Open
Abstract
Tissue perfusion is a necessary condition for vessel survival that can be compromised under ischemic conditions. Following stroke, delayed effects of early brain reperfusion on the vascular substrate necessary for remodeling, perfusion and maintenance of proper peri-lesional hemodynamics are unknown. Such aspects of ischemic injury progression may be critical for neurological recovery in stroke patients. This study aims to describe the impact of early, non-thrombolytic reperfusion on the vascular brain component and its potential contribution to tissue remodeling and long-term functional recovery beyond the acute phase after stroke in 3-month-old male C57bl/6 mice. Permanent (pMCAO) and transient (60 min, tMCAO) brain ischemia mouse models were used for characterizing the effect of early, non-thrombolytic reperfusion on the brain vasculature. Analysis of different vascular parameters (vessel density, proliferation, degeneration and perfusion) revealed that, while early middle cerebral artery recanalization was not sufficient to prevent sub-acute vascular degeneration within the ischemic brain regions, brain reperfusion promoted a secondary wave of vascular remodeling in the peri-lesional regions, which led to improved perfusion of the ischemic boundaries and late-phase neurological recovery. This study concluded that acute, non-thrombolytic artery recanalization following stroke favors late-phase vascular remodeling and improves peri-lesional perfusion, contributing to secondary functional recovery.
Collapse
Affiliation(s)
- Violeta Durán-Laforet
- Unidad de Investigación Neurovascular, Departamento de Farmacología y Toxicología, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - David Fernández-López
- Unidad de Investigación Neurovascular, Departamento de Farmacología y Toxicología, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Alicia García-Culebras
- Unidad de Investigación Neurovascular, Departamento de Farmacología y Toxicología, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Juan González-Hijón
- Unidad de Investigación Neurovascular, Departamento de Farmacología y Toxicología, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Ana Moraga
- Unidad de Investigación Neurovascular, Departamento de Farmacología y Toxicología, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Sara Palma-Tortosa
- Unidad de Investigación Neurovascular, Departamento de Farmacología y Toxicología, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Isaac García-Yébenes
- Unidad de Investigación Neurovascular, Departamento de Farmacología y Toxicología, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Adrián Vega-Pérez
- Unidad de Investigación Neurovascular, Departamento de Farmacología y Toxicología, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Ignacio Lizasoain
- Unidad de Investigación Neurovascular, Departamento de Farmacología y Toxicología, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - María Ángeles Moro
- Unidad de Investigación Neurovascular, Departamento de Farmacología y Toxicología, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| |
Collapse
|
55
|
Mangin G, Cogo A, Moisan A, Bonnin P, Maïer B, Kubis N. Intravenous Administration of Human Adipose Derived-Mesenchymal Stem Cells Is Not Efficient in Diabetic or Hypertensive Mice Subjected to Focal Cerebral Ischemia. Front Neurosci 2019; 13:718. [PMID: 31379478 PMCID: PMC6646672 DOI: 10.3389/fnins.2019.00718] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Accepted: 06/26/2019] [Indexed: 12/14/2022] Open
Abstract
As the second cause of death and cognitive decline in industrialized countries, stroke is a major burden for society. Vascular risks factors such as hypertension and diabetes are involved in most stroke patients, aggravate stroke severity, but are still poorly taken into account in preclinical studies. Microangiopathy and sustained inflammation are exacerbated, likely explaining the severity of stroke in those patients. We sought to demonstrate that intravenous administration of human adipose derived-mesenchymal stem cells (hADMSC) that have immunomodulatory properties, could accelerate sensorimotor recovery, prevent long-term spatial memory impairment and promote neurogenesis, in diabetic or hypertensive mice, subjected to permanent middle cerebral artery occlusion (pMCAo). Diabetic (streptozotocin IP) or hypertensive (L-NAME in drinking water) male C57Bl6 mice subjected to pMCAo, were treated by hADMSC (500,000 cells IV) 2 days after cerebral ischemia induction. Infarct volume, neurogenesis, microglial/macrophage density, T-lymphocytes density, astrocytes density, and vessel density were monitored 7 days after cells injection and at 6 weeks. Neurological sensorimotor deficit and spatial memory were assessed until 6 weeks post-stroke. Whatever the vascular risk factor, hADMSC showed no effect on functional sensorimotor recovery or cognitive decline prevention at short or long-term assessment, nor significantly modified neurogenesis, microglial/macrophage, T-lymphocytes, astrocytes, and vessel density. This work is part of a European program (H2020, RESSTORE). We discuss the discrepancy of our results with those obtained in rats and the optimal cell injection time frame, source and type of cells according to the species stroke model. A comprehensive understanding of the mechanisms preventing recovery should help for successful clinical translation, but first could allow identifying good and bad responders to cell therapy in stroke.
Collapse
Affiliation(s)
| | - Adrien Cogo
- INSERM, U965, CART, Paris, France.,INSERM, U1148, Laboratory for Vascular and Translational Science, Universite de Paris, Paris, France
| | - Anaïck Moisan
- Unité de Thérapie et d'Ingénierie Cellulaire, EFS Auvergne Rhône Alpes, Saint-Ismier, France
| | - Philippe Bonnin
- INSERM, U965, CART, Paris, France.,INSERM, U1148, Laboratory for Vascular and Translational Science, Universite de Paris, Paris, France.,Service de Physiologie Clinique-Explorations Fonctionnelles, AP-HP, Hôpital Lariboisière, Paris, France
| | | | - Nathalie Kubis
- INSERM, U965, CART, Paris, France.,INSERM, U1148, Laboratory for Vascular and Translational Science, Universite de Paris, Paris, France.,Service de Physiologie Clinique-Explorations Fonctionnelles, AP-HP, Hôpital Lariboisière, Paris, France
| | | |
Collapse
|
56
|
Zhan Y, Li MZ, Yang L, Feng XF, Zhang QX, Zhang N, Zhao YY, Zhao H. An MRI Study of Neurovascular Restorative After Combination Treatment With Xiaoshuan Enteric-Coated Capsule and Enriched Environment in Rats After Stroke. Front Neurosci 2019; 13:701. [PMID: 31354412 PMCID: PMC6630081 DOI: 10.3389/fnins.2019.00701] [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: 03/05/2019] [Accepted: 06/20/2019] [Indexed: 12/13/2022] Open
Abstract
Xiaoshuan enteric-coated capsule (XSEC) is a Chinese medicinal compound widely used for treatment of ischemic cerebrovascular diseases. Enriched environment (EE) is an effective rehabilitative protocol designed to enhance sensorimotor, cognitive and social stimulation. This study aimed to apply magnetic resonance imaging (MRI) to non-invasively assess whether EE could augment the therapeutic benefits of XSEC on post-ischemic neurovascular remodeling. Male Sprague–Dawley rats were subjected to permanent middle cerebral artery occlusion (MCAO) and treated with XSEC and EE alone or combination for 30 consecutive days. Beam walking test and Morris water maze (MWM) test were performed to evaluate motor and cognitive function, respectively. Multimodal MRI was applied to examine alterations to brain structures, intracranial vessels, and cerebral perfusion on the 31st day after MCAO. Double-immunofluorescent staining was used to evaluate neurogenesis and angiogenesis. Western blot and RT-PCR were used to detect the expressions of vascular endothelial growth factor (VEGF), angiopoietin-1 (Ang-1), angiopoietin-2 (Ang-2), and the axon guidance molecules. Combination therapy with XSEC and EE significantly reduced cystic volume compared with XSEC and EE monotherapies. In line with this, combination treated rats performed better in the beam walking test and exhibited improved spatial memory in the probe trial of the MWM. Moreover, XSEC and EE combination treatment improved cerebral blood flow (CBF), amplified angiogenesis and upregulated VEGF protein levels. This proangiogenic effect was consistent with the increased progenitor cell proliferation and neuronal differentiation in the peri-infarct cortex and striatum. Specifically, the combined therapy of XSEC and EE markedly increased the Netrin-1 and Robo-1 protein expression levels compared with vehicle group, while no difference was observed between XSEC or EE monotherapy and vehicle group. Together, these findings indicate that the combination of XSEC and EE benefits neurovascular reorganization. This correlates with restoration of CBF, promotion of neurogenesis and angiogenesis, and activation of the intrinsic axonal guidance molecules, thereby facilitating greater physical rehabilitation after ischemic stroke.
Collapse
Affiliation(s)
- Yu Zhan
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| | - Man-Zhong Li
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| | - Le Yang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| | - Xue-Feng Feng
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| | - Qiu-Xia Zhang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| | - Nan Zhang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| | - Yuan-Yuan Zhao
- Medical Imaging Laboratory of Core Facility Center, Capital Medical University, Beijing, China
| | - Hui Zhao
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| |
Collapse
|
57
|
Komori M, Ago T, Wakisaka Y, Nakamura K, Tachibana M, Yoshikawa Y, Shibahara T, Yamanaka K, Kuroda J, Kitazono T. Early initiation of a factor Xa inhibitor can attenuate tissue repair and neurorestoration after middle cerebral artery occlusion. Brain Res 2019; 1718:201-211. [PMID: 31103522 DOI: 10.1016/j.brainres.2019.05.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 05/14/2019] [Accepted: 05/16/2019] [Indexed: 10/26/2022]
Abstract
The timing of anti-coagulation therapy initiation after acute cardioembolic stroke remains controversial. We investigated the effects of post-stroke administration of a factor Xa inhibitor in mice, focusing on tissue repair and functional restoration outcomes. We initiated administration of rivaroxaban, a Xa inhibitor, immediately after permanent distal middle cerebral artery occlusion (pMCAO) in CB-17 mice harboring few leptomeningeal anastomoses at baseline. Rivaroxaban initiated immediately after pMCAO hindered the recovery of blood flow in ischemic areas by inhibiting leptomeningeal anastomosis development, and led to impaired restoration of neurologic functions with less extensive peri-infarct astrogliosis. Within infarct areas, angiogenesis and fibrotic responses were attenuated in rivaroxaban-fed mice. Furthermore, inflammatory responses, including the accumulation of neutrophils and monocytes/macrophages, local secretion of pro-inflammatory cytokines, and breakdown of the blood-brain barrier, were enhanced in infarct areas in mice treated immediately with rivaroxaban following pMCAO. The detrimental effects were not found when rivaroxaban was initiated after transient MCAO or on day 7 after pMCAO. Collectively, early post-stroke initiation of a factor Xa inhibitor may suppress leptomeningeal anastomosis development and blood flow recovery in ischemic areas, thereby resulting in attenuated tissue repair and functional restoration unless occluded large arteries are successfully recanalized.
Collapse
Affiliation(s)
- Motohiro Komori
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Tetsuro Ago
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan.
| | - Yoshinobu Wakisaka
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Kuniyuki Nakamura
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Masaki Tachibana
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yoji Yoshikawa
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Tomoya Shibahara
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Kei Yamanaka
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Junya Kuroda
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Takanari Kitazono
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| |
Collapse
|
58
|
Zhang Q, Zhou M, Wu X, Li Z, Liu B, Gao W, Yue J, Liu T. Promoting therapeutic angiogenesis of focal cerebral ischemia using thrombospondin-4 (TSP4) gene-modified bone marrow stromal cells (BMSCs) in a rat model. J Transl Med 2019; 17:111. [PMID: 30947736 PMCID: PMC6449913 DOI: 10.1186/s12967-019-1845-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 03/11/2019] [Indexed: 01/01/2023] Open
Abstract
Background A stroke caused by angiostenosis always has a poor prognosis. Bone marrow stromal cells (BMSC) are widely applied in vascular regeneration. Recently, thrombospondin-4 (TSP4) was reported to promote the regeneration of blood vessels and enhance the function of endothelial cells in angiogenesis. In this work, we observed the therapeutic effect of TSP4-overexpressing BMSCs on angiogenesis post-stroke. Methods We subcloned the tsp4 gene into a lentivirus expression vector system and harvested the tsp4 lentivirus using 293FT cells. Primary BMSCs were then successfully infected by the tsp4 virus, and overexpression of GFP-fused TSP4 was confirmed by both western blot and immunofluorescence. In vitro, TSP4-overexpressing BMSCs and wild-type BMSCs were co-cultured with human umbilical vein endothelial cells (HUVECs). The expression level of TSP4, vascular endothelial growth factor (VEGF) and transforming growth factor-β (TGF-β) in the supernatant were detected by enzyme-linked immunosorbent assay (ELISA). Wound healing, tube formation and an arterial ring test were performed to estimate the ability of TSP4-overexpressing BMSCs to promote the angiogenesis of endothelial cells. Using a rat permanent middle cerebral artery occlusion (MCAO) model, the effect of TSP4-overexpressing BMSCs on the regeneration of blood vessels was systematically tested by the neurological function score, immunohistochemistry and immunofluorescence staining assays. Results Our results demonstrated that TSP4-overexpressing BMSCs largely increased the expression of VEGF, angiopoietin-1 (Ang-1), matrix metalloprotein 9 (MMP9), matrix metalloprotein 2 (MMP2) and p-Cdc42/Rac1 in endothelial cells. TSP4-BMSC treatment notably up-regulated the TGF-β/Smad2/3 signalling pathway in HUVECs. In vivo, the TSP4-BMSC infusion improved the neurological function score of MCAO rats and expanded the expression of the von Willebrand factor (vWF), Ang-1, MMP2 and MMP9 proteins in cerebral ischemic penumbra. Conclusions Our data illustrate that TSP4-BMSCs can promote the proliferation and migration of endothelial cells and tube formation. We found that TSP4-BMSC infusion can promote the recovery of neural function post-stroke. The tsp4 gene-modified BMSCs provides a better therapeutic effect than that of wild-type BMSCs. Electronic supplementary material The online version of this article (10.1186/s12967-019-1845-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Biotherapy and Oncology, Shenzhen Luohu People's Hospital, Shenzhen, 518001, Guangdong, People's Republic of China.,Public Service Platform for Cell Quality Testing and Evaluation of Shenzhen, Shenzhen, 518001, Guangdong, People's Republic of China
| | - Meiling Zhou
- Department of Biotherapy and Oncology, Shenzhen Luohu People's Hospital, Shenzhen, 518001, Guangdong, People's Republic of China.,Public Service Platform for Cell Quality Testing and Evaluation of Shenzhen, Shenzhen, 518001, Guangdong, People's Republic of China
| | - Xiangfeng Wu
- Department of Biotherapy and Oncology, Shenzhen Luohu People's Hospital, Shenzhen, 518001, Guangdong, People's Republic of China.,Mudanjiang Medical University, Mudanjiang, 157011, Heilongjiang, People's Republic of China
| | - Zhu Li
- Department of Biotherapy and Oncology, Shenzhen Luohu People's Hospital, Shenzhen, 518001, Guangdong, People's Republic of China.,Public Service Platform for Cell Quality Testing and Evaluation of Shenzhen, Shenzhen, 518001, Guangdong, People's Republic of China
| | - Bing Liu
- Department of Biotherapy and Oncology, Shenzhen Luohu People's Hospital, Shenzhen, 518001, Guangdong, People's Republic of China.,Public Service Platform for Cell Quality Testing and Evaluation of Shenzhen, Shenzhen, 518001, Guangdong, People's Republic of China
| | - Wenbin Gao
- Department of Biotherapy and Oncology, Shenzhen Luohu People's Hospital, Shenzhen, 518001, Guangdong, People's Republic of China
| | - Jin Yue
- The 230th Hospital of the Chinese PLA, Dandong, Liaoning, People's Republic of China.
| | - Tao Liu
- Department of Biotherapy and Oncology, Shenzhen Luohu People's Hospital, Shenzhen, 518001, Guangdong, People's Republic of China. .,Public Service Platform for Cell Quality Testing and Evaluation of Shenzhen, Shenzhen, 518001, Guangdong, People's Republic of China.
| |
Collapse
|
59
|
Venkat P, Chen J, Chopp M. Exosome-mediated amplification of endogenous brain repair mechanisms and brain and systemic organ interaction in modulating neurological outcome after stroke. J Cereb Blood Flow Metab 2018; 38:2165-2178. [PMID: 29888985 PMCID: PMC6282218 DOI: 10.1177/0271678x18782789] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Ischemic stroke is caused by a regional interruption of cerebral blood flow to the brain. Rigorous pre-clinical and clinical research has made landmark progress in stroke treatment using thrombolytics and endovascular thrombectomy. Although numerous successful neuroprotective therapeutic agents for ischemic stroke have been reported in pre-clinical studies, most of them failed in clinical testing. Persistent pre-clinical research has demonstrated that the ischemic brain is not only passively dying but is also actively recovering. Within the neurovascular niche in the peri-infarct tissue, repair mechanisms thrive on the interactions between the neural and vascular compartments. In this review, we discuss exogenous therapy using mesenchymal stromal cell-derived exosomes to amplify endogenous brain repair mechanisms and to induce neurorestorative effects after stroke. Emerging evidence indicates that multiple communication axes between the various organs such as the brain, heart, kidney and gut, and whole body immune response mediated by the spleen can also affect stroke outcome. Therefore, in this review, we summarize this evidence and initiate a discussion on the potential to improve stroke outcome by amplifying multiple brain repair mechanisms after stroke, and by targeting peripheral organs and downstream events to enhance recovery in the injured brain and promote over all well being.
Collapse
Affiliation(s)
- Poornima Venkat
- 1 Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Jieli Chen
- 1 Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Michael Chopp
- 1 Department of Neurology, Henry Ford Hospital, Detroit, MI, USA.,2 Department of Physics, Oakland University, Rochester, MI, USA
| |
Collapse
|
60
|
Venkat P, Yan T, Chopp M, Zacharek A, Ning R, Van Slyke P, Dumont D, Landschoot-Ward J, Liang L, Chen J. Angiopoietin-1 Mimetic Peptide Promotes Neuroprotection after Stroke in Type 1 Diabetic Rats. Cell Transplant 2018; 27:1744-1752. [PMID: 30124060 PMCID: PMC6300775 DOI: 10.1177/0963689718791568] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 05/24/2018] [Accepted: 06/27/2018] [Indexed: 12/02/2022] Open
Abstract
Angiopoietin-1 (Ang1) mediates vascular maturation and immune response. Diabetes decreases Ang1 expression and disrupts Ang1/Tie2 signaling activity. Vasculotide is an Ang1 mimetic peptide, and has anti-inflammatory effects. In this study, we test the hypothesis that vasculotide treatment induces neuroprotection and decreases inflammation after stroke in type 1 diabetic (T1DM) rats. T1DM rats were subjected to embolic middle cerebral artery occlusion (MCAo) and treated with: 1) phosphate buffered saline (PBS); 2) vasculotide (3µg/kg, i.p. injection) administered half an hour prior to MCAo and at 8 and 24 hours after MCAo. Rats were sacrificed at 48 h after MCAo. Neurological function, infarct volume, hemorrhage, blood brain barrier (BBB) permeability and neuroinflammation were measured. Vasculotide treatment of T1DM-MCAo rats significantly improves functional outcome, decreases infarct volume and BBB permeability, but does not decrease brain hemorrhagic transformation compared with PBS-treated T1DM-MCAo rats. In the ischemic brain, Vasculotide treatment significantly decreases apoptosis, number of cleaved-caspase-3 positive cells, the expression of monocyte chemotactic protein-1 (MCP-1) and tumor necrosis factor (TNF-α). Western blot analysis shows that vasculotide significantly decreases expression of receptor for advanced glycation end products (RAGE), MCP-1 and TNF-α in the ischemic brain compared with T1DM-MCAo rats. Vasculotide treatment in cultured primary cortical neurons (PCN) significantly decreases TLR4 expression compared with control. Decreased neuroinflammation and reduced BBB leakage may contribute, at least in part, to vasculotide-induced neuroprotective effects after stroke in T1DM rats.
Collapse
Affiliation(s)
- Poornima Venkat
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Tao Yan
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
- Department of Physics, Oakland University, Rochester, MI, USA
| | - Alex Zacharek
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Ruizhuo Ning
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | | | - Daniel Dumont
- Department of Medical Biophysics, Sunnybrook Research Institute, University of Toronto, Canada
| | | | - Linlin Liang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
- Reproductive Medical Center, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Jieli Chen
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| |
Collapse
|
61
|
Vogel A, Upadhya R, Shetty AK. Neural stem cell derived extracellular vesicles: Attributes and prospects for treating neurodegenerative disorders. EBioMedicine 2018; 38:273-282. [PMID: 30472088 PMCID: PMC6306394 DOI: 10.1016/j.ebiom.2018.11.026] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 11/04/2018] [Accepted: 11/14/2018] [Indexed: 02/06/2023] Open
Abstract
Neural stem cell (NSC) grafting in conditions such as aging, brain injury, and neurodegenerative diseases promotes regeneration, plasticity and functional recovery. Recent studies have revealed that administration of NSC-derived extracellular vesicles (NSC-EVs) via non-invasive approaches can also afford therapeutic benefits. This review confers the properties and therapeutic promise of EVs secreted by NSCs. NSC-EVs enriched with specific miRNAs mediate multiple functions in physiological and pathological conditions, which include modulation of the proximate microenvironment, facilitating the entry of viruses into cells, functioning as independent metabolic units, operating as a microglial morphogen and influencing the diverse aspects of brain function in adulthood including the process of aging. Due to their anti-inflammatory, neurogenic and neurotrophic effects, NSC-EVs are also useful for treating multiple neurodegenerative diseases. Although only a few studies have demonstrated the efficacy of NSC-EVs to treat brain impairments, the promise is enormous. Moving forward, the use of well-characterized NSC-EVs generated in specific culture conditions and NSC-EVs that are engineered to carry the desired miRNAs, mRNAs and proteins have great promise for treating brain injury and neurogenerative diseases. Notably, the possibility of targeting NSC-EVs to specific neuronal types or brain regions would enable managing of diverse neurodegenerative conditions with minimal side effects.
Collapse
Affiliation(s)
- Andrew Vogel
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center College of Medicine, 1114 TAMU, 206 Olsen Boulevard, College Station, Texas, United States
| | - Raghavendra Upadhya
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center College of Medicine, 1114 TAMU, 206 Olsen Boulevard, College Station, Texas, United States; Olin E. Teague Veterans' Medical Center, Central Texas Veterans Health Care System, Temple, TX, United States
| | - Ashok K Shetty
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center College of Medicine, 1114 TAMU, 206 Olsen Boulevard, College Station, Texas, United States; Olin E. Teague Veterans' Medical Center, Central Texas Veterans Health Care System, Temple, TX, United States.
| |
Collapse
|
62
|
Integrin α5β1-Ang1/Tie2 receptor cross-talk regulates brain endothelial cell responses following cerebral ischemia. Exp Mol Med 2018; 50:1-12. [PMID: 30185785 PMCID: PMC6123805 DOI: 10.1038/s12276-018-0145-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 04/06/2018] [Accepted: 05/14/2018] [Indexed: 11/08/2022] Open
Abstract
We have previously demonstrated that in response to cerebral ischemia (CI), the growth factor angiopoietin-1 (Ang1) and α5β1 integrin are both induced in cerebral vessels, which likely provide positive signals driving the endogenous angiogenic response and vascular protection after CI. However, the precise relationship between endothelial Ang1 and α5β1 integrin after CI remains poorly understood. Here, we investigated the effects of the interaction between the Ang1/Tie2 system and α5β1 integrin on brain endothelial cells (BECs) under cerebral ischemic conditions in vivo and in vitro. Immunofluorescence analysis demonstrated that integrin α5β1 co-localized with Tie2/phosphorylated Tie2 on cerebral vessels in the penumbra. The in vitro study showed that oxygen-glucose deprivation/restoration (OGD/R) induced the expression of the Ang1 receptor Tie2 on BECs in a manner similar to that for integrin α5 and Ang1 in response to OGD/R, accompanied by increased activation of Tie2 and its downstream effectors focal adhesion kinase (FAK) and Akt. Knockdown of α5 integrin markedly suppressed OGD/R-induced Tie2 receptor activation in BECs, while in contrast, priming BECs with Ang1 promoted the expression of α5 integrin as well as the Tie2 downstream transcription factor Ets-1 in OGD-treated BECs. In line with this, Ets-1 knockdown significantly attenuated Ang1-mediated upregulation of α5 integrin. Functionally, Ang1 induced cell migration and tube formation of BECs after OGD, but this effect was inhibited by diminishment of the levels of α5 integrin in BECs. Taken together, our data indicate that the Ang1/Tie2 system cross-talks with integrin α5β1 in BECs after CI, which may contribute to the endogenous angiogenic vascular protective response following CI.
Collapse
|
63
|
Sun C, Yang F, Wang C, Wang Z, Zhang Y, Ming D, Du J. Mutual Information-Based Brain Network Analysis in Post-stroke Patients With Different Levels of Depression. Front Hum Neurosci 2018; 12:285. [PMID: 30065639 PMCID: PMC6056615 DOI: 10.3389/fnhum.2018.00285] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 06/25/2018] [Indexed: 11/15/2022] Open
Abstract
Post-stroke depression (PSD) is the most common stroke-related emotional disorder, and it severely affects the recovery process. However, more than half cases are not correctly diagnosed. This study was designed to develop a new method to assess PSD using EEG signal to analyze the specificity of PSD patients' brain network. We have 107 subjects attended in this study (72 stabilized stroke survivors and 35 non-depressed healthy subjects). A Hamilton Depression Rating Scale (HDRS) score was determined for all subjects before EEG data collection. According to HDRS score, the 72 patients were divided into 3 groups: post-stroke non-depression (PSND), post-stroke mild depression (PSMD) and post-stroke depression (PSD). Mutual information (MI)-based graph theory was used to analyze brain network connectivity. Statistical analysis of brain network characteristics was made with a threshold of 10-30% of the strongest MIs. The results showed significant weakened interhemispheric connections and lower clustering coefficient in post-stroke depressed patients compared to those in healthy controls. Stroke patients showed a decreasing trend in the connection between the parietal-occipital and the frontal area as the severity of the depression increased. PSD subjects showed abnormal brain network connectivity and network features based on EEG, suggesting that MI-based brain network may have the potential to assess the severity of depression post stroke.
Collapse
Affiliation(s)
- Changcheng Sun
- Rehabilitation Medical Department, Tianjin Union Medical Centre, Tianjin, China
| | - Fei Yang
- Department of Health and Exercise Science, Tianjin University of Sport, Tianjin, China
| | - Chunfang Wang
- Rehabilitation Medical Department, Tianjin Union Medical Centre, Tianjin, China
| | - Zhonghan Wang
- Rehabilitation Medical Department, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ying Zhang
- Rehabilitation Medical Department, Tianjin Union Medical Centre, Tianjin, China
| | - Dong Ming
- Department of Biomedical Engineering, College of Precision Instrument and Opto-Electronics Engineering, Tianjin University, Tianjin, China
| | - Jingang Du
- Rehabilitation Medical Department, Tianjin Union Medical Centre, Tianjin, China
| |
Collapse
|
64
|
Li MZ, Zhang Y, Zou HY, Ouyang JY, Zhan Y, Yang L, Cheng BCY, Wang L, Zhang QX, Lei JF, Zhao YY, Zhao H. Investigation of Ginkgo biloba extract (EGb 761) promotes neurovascular restoration and axonal remodeling after embolic stroke in rat using magnetic resonance imaging and histopathological analysis. Biomed Pharmacother 2018; 103:989-1001. [DOI: 10.1016/j.biopha.2018.04.125] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 04/14/2018] [Accepted: 04/17/2018] [Indexed: 02/06/2023] Open
|
65
|
Yan T, Venkat P, Chopp M, Zacharek A, Yu P, Ning R, Qiao X, Kelley MR, Chen J. APX3330 Promotes Neurorestorative Effects after Stroke in Type One Diabetic Rats. Aging Dis 2018; 9:453-466. [PMID: 29896433 PMCID: PMC5988600 DOI: 10.14336/ad.2017.1130] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 11/30/2017] [Indexed: 12/15/2022] Open
Abstract
APX3330 is a selective inhibitor of APE1/Ref-1 redox activity. In this study, we investigate the therapeutic effects and underlying mechanisms of APX3330 treatment in type one diabetes mellitus (T1DM) stroke rats. Adult male Wistar rats were induced with T1DM and subjected to transient middle cerebral artery occlusion (MCAo) and treated with either PBS or APX3330 (10mg/kg, oral gavage) starting at 24h after MCAo, and daily for 14 days. Rats were sacrificed at 14 days after MCAo and, blood brain barrier (BBB) permeability, ischemic lesion volume, immunohistochemistry, cell death assay, Western blot, real time PCR, and angiogenic ELISA array were performed. Compared to PBS treatment, APX3330 treatment of stroke in T1DM rats significantly improves neurological functional outcome, decreases lesion volume, and improves BBB integrity as well as decreases total vessel density and VEGF expression, while significantly increases arterial density in the ischemic border zone (IBZ). APX3330 significantly increases myelin density, oligodendrocyte number, oligodendrocyte progenitor cell number, synaptic protein expression, and induces M2 macrophage polarization in the IBZ of T1DM stroke rats. Compared to PBS treatment, APX3330 treatment significantly decreases plasminogen activator inhibitor type-1 (PAI-1), monocyte chemotactic protein-1 and matrix metalloproteinase 9 (MMP9) and receptor for advanced glycation endproducts expression in the ischemic brain of T1DM stroke rats. APX3330 treatment significantly decreases cell death and MMP9 and PAI-1 gene expression in cultured primary cortical neurons subjected to high glucose and oxygen glucose deprivation, compared to untreated control cells. APX3330 treatment increases M2 macrophage polarization and decreases inflammatory factor expression in the ischemic brain as well as promotes neuroprotective and neurorestorative effects after stroke in T1DM rats.
Collapse
Affiliation(s)
- Tao Yan
- 1Gerontology Institute, Neurology, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China.,2Department of Neurology, Henry Ford hospital, Detroit, MI, USA
| | - Poornima Venkat
- 2Department of Neurology, Henry Ford hospital, Detroit, MI, USA
| | - Michael Chopp
- 2Department of Neurology, Henry Ford hospital, Detroit, MI, USA.,3Department of Physics, Oakland University, Rochester, MI, USA
| | - Alex Zacharek
- 2Department of Neurology, Henry Ford hospital, Detroit, MI, USA
| | - Peng Yu
- 2Department of Neurology, Henry Ford hospital, Detroit, MI, USA
| | - Ruizhuo Ning
- 2Department of Neurology, Henry Ford hospital, Detroit, MI, USA.,4Department of Neurology, First Hospital Harbin, Harbin, China
| | - Xiaoxi Qiao
- 5Department of Ophthalmology, Henry Ford Hospital, Detroit, MI, USA
| | - Mark R Kelley
- 6Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jieli Chen
- 2Department of Neurology, Henry Ford hospital, Detroit, MI, USA
| |
Collapse
|
66
|
Menet R, Bernard M, ElAli A. Hyperlipidemia in Stroke Pathobiology and Therapy: Insights and Perspectives. Front Physiol 2018; 9:488. [PMID: 29867540 PMCID: PMC5962805 DOI: 10.3389/fphys.2018.00488] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 04/17/2018] [Indexed: 12/31/2022] Open
Affiliation(s)
- Romain Menet
- Neuroscience Axis, CHU de Québec Research Center (CHUL), Québec City, QC, Canada.,Department of Psychiatry and Neuroscience, Faculty of Medicine, Laval University, Québec City, QC, Canada
| | - Maxime Bernard
- Neuroscience Axis, CHU de Québec Research Center (CHUL), Québec City, QC, Canada.,Department of Psychiatry and Neuroscience, Faculty of Medicine, Laval University, Québec City, QC, Canada
| | - Ayman ElAli
- Neuroscience Axis, CHU de Québec Research Center (CHUL), Québec City, QC, Canada.,Department of Psychiatry and Neuroscience, Faculty of Medicine, Laval University, Québec City, QC, Canada
| |
Collapse
|
67
|
Chen J, Chopp M. Exosome Therapy for Stroke. Stroke 2018; 49:1083-1090. [PMID: 29669873 PMCID: PMC6028936 DOI: 10.1161/strokeaha.117.018292] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 08/28/2017] [Accepted: 08/30/2017] [Indexed: 12/11/2022]
Affiliation(s)
- Jieli Chen
- From the Department of Neurology, Henry Ford Hospital, Detroit, MI (J.C., M.C.)
- Department of Geriatrics, Tianjin Medical University General Hospital, China (J.C.)
- Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education, China (J.C.)
| | - Michael Chopp
- From the Department of Neurology, Henry Ford Hospital, Detroit, MI (J.C., M.C.)
- Department of Physics, Oakland University, Rochester, MI (M.C.)
| |
Collapse
|
68
|
Kaur H, Sarmah D, Saraf J, Vats K, Kalia K, Borah A, Yavagal DR, Dave KR, Ghosh Z, Bhattacharya P. Noncoding RNAs in ischemic stroke: time to translate. Ann N Y Acad Sci 2018; 1421:19-36. [DOI: 10.1111/nyas.13612] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 12/11/2017] [Accepted: 01/08/2018] [Indexed: 12/21/2022]
Affiliation(s)
- Harpreet Kaur
- Department of Pharmacology and Toxicology; National Institute of Pharmaceutical Education and Research (NIPER); Ahmedabad, Gandhinagar Gujarat India
| | - Deepaneeta Sarmah
- Department of Pharmacology and Toxicology; National Institute of Pharmaceutical Education and Research (NIPER); Ahmedabad, Gandhinagar Gujarat India
| | - Jackson Saraf
- Department of Pharmacology and Toxicology; National Institute of Pharmaceutical Education and Research (NIPER); Ahmedabad, Gandhinagar Gujarat India
| | - Kanchan Vats
- Department of Pharmacology and Toxicology; National Institute of Pharmaceutical Education and Research (NIPER); Ahmedabad, Gandhinagar Gujarat India
| | - Kiran Kalia
- Department of Pharmacology and Toxicology; National Institute of Pharmaceutical Education and Research (NIPER); Ahmedabad, Gandhinagar Gujarat India
| | - Anupom Borah
- Cellular and Molecular Neurobiology Laboratory; Department of Life Science and Bioinformatics; Assam University; Silchar Assam India
| | - Dileep R. Yavagal
- Department of Neurology and Neurosurgery; University of Miami Miller School of Medicine; Miami Florida
| | - Kunjan R. Dave
- Department of Neurology and Neurosurgery; University of Miami Miller School of Medicine; Miami Florida
| | - Zhumur Ghosh
- Department of Bioinformatics; Bose Institute; Kolkata India
| | - Pallab Bhattacharya
- Department of Pharmacology and Toxicology; National Institute of Pharmaceutical Education and Research (NIPER); Ahmedabad, Gandhinagar Gujarat India
- Department of Neurosurgery, Boston Children's Hospital; Harvard Medical School; Boston Massachusetts
| |
Collapse
|
69
|
Venkat P, Chopp M, Chen J. Cell-Based and Exosome Therapy in Diabetic Stroke. Stem Cells Transl Med 2018; 7:451-455. [PMID: 29498242 PMCID: PMC5980126 DOI: 10.1002/sctm.18-0014] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 02/15/2018] [Indexed: 12/21/2022] Open
Abstract
Stroke is a global health concern and it is imperative that therapeutic strategies with wide treatment time frames be developed to improve neurological outcome in patients. Patients with diabetes mellitus who suffer a stroke have worse neurological outcomes and long‐term functional recovery than nondiabetic stroke patients. Diabetes induced vascular damage and enhanced inflammatory milieu likely contributes to worse post stroke outcomes. Diabetic stroke patients have an aggravated pathological cascade, and treatments that benefit nondiabetic stroke patients do not necessarily translate to diabetic stroke patients. Therefore, there is a critical need to develop therapeutics for stroke specifically in the diabetic population. Stem cell based therapy for stroke is an emerging treatment option with wide therapeutic time window. Cell‐based therapies for stroke promote endogenous central nervous system repair and neurorestorative mechanisms such as angiogenesis, neurogenesis, vascular remodeling, white matter remodeling, and also modulate inflammatory and immune responses at the local and systemic level. Emerging evidence suggests that exosomes and their cargo microRNA mediate cell therapy derived neurorestorative effects. Exosomes are small vesicles containing protein and RNA characteristic of its parent cell. Exosomes are transported by biological fluids and facilitate communication between neighboring and remote cells. MicroRNAs, a class of naturally occurring, small noncoding RNA sequences, contained within exosomes can regulate recipient cell's signaling pathways and alter protein expression either acting alone or in concert with other microRNAs. In this perspective article, we summarize current knowledge and highlight the promising future of cell based and exosome therapy for stroke and specifically for diabetic stroke. stemcellstranslationalmedicine2018;7:451–455
Collapse
Affiliation(s)
| | - Michael Chopp
- Neurology Henry Ford Hospital, Detroit, Michigan, USA.,Department of Physics, Oakland University, Rochester, Michigan, USA
| | - Jieli Chen
- Neurology Henry Ford Hospital, Detroit, Michigan, USA.,Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, People's Republic of China
| |
Collapse
|
70
|
Wei ZZ, Zhang JY, Taylor TM, Gu X, Zhao Y, Wei L. Neuroprotective and regenerative roles of intranasal Wnt-3a administration after focal ischemic stroke in mice. J Cereb Blood Flow Metab 2018; 38:404-421. [PMID: 28430000 PMCID: PMC5851145 DOI: 10.1177/0271678x17702669] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 02/27/2017] [Accepted: 03/01/2017] [Indexed: 01/31/2023]
Abstract
Wnt signaling is a conserved pathway involved in expansion of neural progenitors and lineage specification during development. However, the role of Wnt signaling in the post-stroke brain has not been well-elucidated. We hypothesized that Wnt-3a would play an important role for neurogenesis and brain repair. Adult male mice were subjected to a focal ischemic stroke targeting the sensorimotor cortex. Mice that received Wnt-3a (2 µg/kg/day, 1 h after stroke and once a day for the next 2 days, intranasal delivery) had reduced infarct volume compared to stroke controls. Wnt-3a intranasal treatment of seven days upregulated the expression of brain-derived growth factor (BDNF), increased the proliferation and migration of neuroblasts from the subventricular zone (SVZ), resulting in increased numbers of newly formed neurons and endothelial cells in the peri-infarct zone. Both the molecular and cellular effects of Wnt-3a were blocked by the Wnt specific inhibitors XAV-939 or Dkk-1. In functional assays, Wnt-3a treatment enhanced the local cerebral blood flow (LCBF) in the peri-infarct, as well as improved sensorimotor functions in a battery of behavioral tests. Together, our data demonstrates that the Wnt-3a signaling can act as a dual neuroprotective and regenerative factor for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Zheng Zachory Wei
- Laboratories of Stem Cell Biology and Regenerative Medicine, Experimental Research Center and Neurological Disease Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - James Ya Zhang
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Tammi M Taylor
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Xiaohuan Gu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Yingying Zhao
- Laboratories of Stem Cell Biology and Regenerative Medicine, Experimental Research Center and Neurological Disease Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Ling Wei
- Laboratories of Stem Cell Biology and Regenerative Medicine, Experimental Research Center and Neurological Disease Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
71
|
Farajdokht F, Farhoudi M, Majdi A, Zamanlu M, Sadigh-Eteghad S, Vahedi S, Mahmoudi J. Testosterone May Hold Therapeutic Promise for the Treatment of Ischemic Stroke in Aging: A Closer Look at Laboratory Findings. Adv Pharm Bull 2018; 9:48-55. [PMID: 31011557 PMCID: PMC6468219 DOI: 10.15171/apb.2019.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 12/20/2018] [Accepted: 12/22/2018] [Indexed: 12/22/2022] Open
Abstract
Male sex is more prone to cerebrovascular disorders, yet the exact role of androgens in cerebral
ischemia remains unclear. Here we reviewed current understanding of testosterone (TES)
neuroprotective activity against ischemic stroke and mechanisms underlying these effects in
aging. TES may exert a neuroprotective effect in aging through pathways including inhibition of
oxidant molecules production, enhancing the enzymatic antioxidant capacity of the brain and
modulation of apoptotic cell death. Given this, a better understanding of the neuroprotective
roles of TES may propose an effective therapeutic strategy to improve the quality of life and
decrease androgen-related cerebrovascular problems in the aging men.
Collapse
Affiliation(s)
- Fereshteh Farajdokht
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Farhoudi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Majdi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masumeh Zamanlu
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Sadigh-Eteghad
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shabnam Vahedi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javad Mahmoudi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
72
|
Shin JA, Kim YA, Kim HW, Kim HS, Lee KE, Kang JL, Park EM. Iron released from reactive microglia by noggin improves myelin repair in the ischemic brain. Neuropharmacology 2018; 133:202-215. [PMID: 29407213 DOI: 10.1016/j.neuropharm.2018.01.038] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 01/21/2018] [Accepted: 01/25/2018] [Indexed: 12/31/2022]
Abstract
We previously reported that the bone morphogenetic protein (BMP) antagonist, noggin, improved the repair process with an increase in the reactive microglia/macrophage population in the ischemic brain. Since BMP plays a role in intracellular iron homeostasis via the hepcidin/ferroportin axis, and iron is required for myelination, this study was aimed to determine whether noggin affected iron status and remyelination in the brain following ischemic stroke. We further examined the effect of blocking the BMP/hepcidin pathway on reactive microglia (BV2) and myelination of oligodendroglial cells (MO3.13) to define the link between microglial iron status and myelin formation. Following the noggin infusion into the ischemic brain of mice, the induction of hepcidin and ferritin protein levels decreased, and the number of myelinated axons and myelin thickness increased at 8 weeks after ischemic stroke. Noggin repressed the increase in hepcidin and ferritin levels in BV2 exposed to lipopolysaccharide (LPS) and oxygen/glucose deprivation and reperfusion (OGD/R). When MO3.13 were exposed to the conditioned media from noggin-treated BV2 (noggin CM) during reperfusion, OGD/R-induced MO3.13 cell death was reduced. Under normal conditions, noggin CM induced myelin production with an increase in ferritin levels in MO3.13, which was reversed by the iron chelator, deferoxamine. These results indicated that noggin altered the iron status in reactive microglia from the iron-storing to the iron-releasing phenotype, which contributed to myelin synthesis by providing iron. We suggest that the BMP/hepcidin pathway can be a target for the regulation of the iron status in microglia to enhance remyelination in the ischemic brain.
Collapse
Affiliation(s)
- Jin A Shin
- Department of Pharmacology, College of Medicine, Ewha Womans University, Seoul, 07985, Republic of Korea; Tissue Injury Defense Research Center, College of Medicine, Ewha Womans University, Seoul, 07985, Republic of Korea
| | - Yul A Kim
- Department of Pharmacology, College of Medicine, Ewha Womans University, Seoul, 07985, Republic of Korea
| | - Hye Won Kim
- Department of Pharmacology, College of Medicine, Ewha Womans University, Seoul, 07985, Republic of Korea; Tissue Injury Defense Research Center, College of Medicine, Ewha Womans University, Seoul, 07985, Republic of Korea
| | - Hee-Sun Kim
- Tissue Injury Defense Research Center, College of Medicine, Ewha Womans University, Seoul, 07985, Republic of Korea; Department of Molecular Medicine, College of Medicine, Ewha Womans University, Seoul, 07985, Republic of Korea
| | - Kyung-Eun Lee
- Department of Pharmacology, College of Medicine, Ewha Womans University, Seoul, 07985, Republic of Korea
| | - Jihee Lee Kang
- Tissue Injury Defense Research Center, College of Medicine, Ewha Womans University, Seoul, 07985, Republic of Korea; Department of Physiology, College of Medicine, Ewha Womans University, Seoul, 07985, Republic of Korea
| | - Eun-Mi Park
- Department of Pharmacology, College of Medicine, Ewha Womans University, Seoul, 07985, Republic of Korea; Tissue Injury Defense Research Center, College of Medicine, Ewha Womans University, Seoul, 07985, Republic of Korea.
| |
Collapse
|
73
|
Han J, Zhang JZ, Zhong ZF, Li ZF, Pang WS, Hu J, Chen LD. Gualou Guizhi decoction promotes neurological functional recovery and neurogenesis following focal cerebral ischemia/reperfusion. Neural Regen Res 2018; 13:1408-1416. [PMID: 30106053 PMCID: PMC6108212 DOI: 10.4103/1673-5374.235296] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Recovery following stroke involves neurogenesis and axonal remodeling within the ischemic brain. Gualou Guizhi decoction (GLGZD) is a Chinese traditional medicine used for the treatment of post-stroke limb spasm. GLGZD has been reported to have neuroprotective effects in cerebral ischemic injury. However, the effects of GLGZD on neurogenesis and axonal remodeling following cerebral ischemia remain unknown. In this study, a rat model of focal cerebral ischemia/reperfusion was established by middle cerebral artery occlusion. Neurological function was assessed immediately after reperfusion using Longa's 5-point scoring system. The rats were randomly divided into vehicle and GLGZD groups. Rats in the sham group were given sham operation. The rats in the GLGZD group were intragastrically administered GLGZD, once daily, for 14 consecutive days. The rats in the vehicle and sham groups were intragastrically administered distilled water. Modified neurological severity score test, balance beam test and foot fault test were used to assess motor functional changes. Nissl staining was performed to evaluate histopathological changes in the brain. Immunofluorescence staining was used to examine cell proliferation using the marker 5-bromo-2′-deoxyuridine (BrdU) as well as expression of the neural precursor marker doublecortin (DCX), the astrocyte marker glial fibrillary acidic protein (GFAP) and the axon regeneration marker growth associated protein-43 (GAP-43). GLGZD substantially mitigated pathological injury, increased the number of BrdU, DCX and GFAP-immunoreactive cells in the subventricular zone of the ischemic hemisphere, increased GAP-43 expression in the cortical peri-infarct region, and improved motor function. These findings suggest that GLGZD promotes neurological functional recovery by increasing cell proliferation, enhancing axonal regeneration, and increasing the numbers of neuronal precursors and astrocytes in the peri-infarct area.
Collapse
Affiliation(s)
- Jing Han
- Institute of Materia Medica, Fujian Academy of Traditional Chinese Medicine, Fuzhou, Fujian Province, China
| | - Ji-Zhou Zhang
- Institute of Materia Medica, Fujian Academy of Traditional Chinese Medicine, Fuzhou, Fujian Province, China
| | - Zhi-Feng Zhong
- Institute of Materia Medica, Fujian Academy of Traditional Chinese Medicine, Fuzhou, Fujian Province, China
| | - Zuan-Fang Li
- Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, China
| | - Wen-Sheng Pang
- Fujian University of Traditional Chinese Medicine; The Second People's Hospital of Fujian Province, Fuzhou, Fujian Province, China
| | - Juan Hu
- Institute of Materia Medica, Fujian Academy of Traditional Chinese Medicine; Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, China
| | - Li-Dian Chen
- Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, China
| |
Collapse
|
74
|
Hu R, Cao Q, Sun Z, Chen J, Zheng Q, Xiao F. A novel method of neural differentiation of PC12 cells by using Opti-MEM as a basic induction medium. Int J Mol Med 2018; 41:195-201. [PMID: 29115371 PMCID: PMC5746309 DOI: 10.3892/ijmm.2017.3195] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 10/11/2017] [Indexed: 12/21/2022] Open
Abstract
The PC12 cell line is a classical neuronal cell model due to its ability to acquire the sympathetic neurons features when deal with nerve growth factor (NGF). In the present study, the authors used a variety of different methods to induce PC12 cells, such as Opti-MEM medium containing different concentrations of fetal bovine serum (FBS) and horse serum compared with RPMI-1640 medium, and then observed the neurite length, differentiation, adhesion, cell proliferation and action potential, as well as the protein levels of axonal growth-associated protein 43 (GAP-43) and synaptic protein synapsin-1, among other differences. Compared with the conventional RPMI-1640 medium induction method, the new approach significantly improved the neurite length of induced cells (2.7 times longer), differentiation rate (30% increase), adhesion rate (21% increase) and expression of GAP-43 and synapsin-1 (three times), as well as reduced cell proliferation. The morphology of induced cells in Opti-MEM medium containing 0.5% FBS was more like that of neurons. Additionally, induced cells were also able to motivate the action potential after treatment for 6 days. Therefore, the research provided a novel, improved induction method of neural differentiation of PC12 cells using Opti-MEM medium containing 0.5% FBS, resulting in a better neuronal model cell line that can be widely used in neurobiology and neuropharmacology research.
Collapse
Affiliation(s)
- Rendong Hu
- Department of Pharmacology, School of Medicine, Jinan University
| | - Qiaoyu Cao
- College of Pharmacy, Jinan University, Guangzhou, Guangdong 510632
| | - Zhongqing Sun
- Department of Anesthesia and Intensive Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong 999077, SAR
| | - Jinying Chen
- Department of Ophthalmology, The First Clinical Medical College of Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Qing Zheng
- College of Pharmacy, Jinan University, Guangzhou, Guangdong 510632
| | - Fei Xiao
- Department of Pharmacology, School of Medicine, Jinan University
| |
Collapse
|
75
|
Okyere B, Creasey M, Lebovitz Y, Theus MH. Temporal remodeling of pial collaterals and functional deficits in a murine model of ischemic stroke. J Neurosci Methods 2018; 293:86-96. [PMID: 28935424 PMCID: PMC5749401 DOI: 10.1016/j.jneumeth.2017.09.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 09/14/2017] [Accepted: 09/16/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND Leptomeningeal anastomoses play a critical role in regulating reperfusion following cerebrovascular obstruction; however, methods to evaluate their temporospatial remodeling remains under investigation. NEW METHOD We combined arteriole-specific vessel painting with histological evaluation to assess the density and diameter of inter-collateral vessels between the middle cerebral artery and anterior cerebral artery (MCA-ACA) or posterior cerebral artery (MCA-PCA) in a murine model of permanent middle cerebral artery occlusion (pMCAO). RESULTS While the overall density was not influenced by pMCAO, the size of MCA-ACA and MCA-PCA vessels had significantly increased 2days post-pMCAO and peaked by 4days compared to the un-injured hemisphere. Using a combination of vessel painting and immunofluorescence, we uniquely observed an induction of cellular division and a remodeling of the smooth muscle cells within the collateral niche following post-pMCAO on whole mount tissue sections. Vessel painting was also applied to pMCAO-injured Cx3cr1GFP mice, in order to identify the spatial relationship between Cx3cr1-positive peripheral-derived monocyte/macrophages and the vessel painted collaterals. Our histological findings were supplemented with analysis of cerebral blood flow using laser Doppler imaging and behavioral changes following pMCAO. COMPARISON WITH EXISTING METHODS Compared to polyurethane and latex methods for collateral labeling, this new method provides detailed cell-type specific analysis within the collateral niche at the microscopic level, which has previously been unavailable. CONCLUSIONS This simple and reproducible combination of techniques is the first to dissect the temporospatial remodeling of pial collateral arterioles. The method will advance investigations into the underlying mechanisms governing the intricate processes of arteriogenesis.
Collapse
Affiliation(s)
- Benjamin Okyere
- The Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, 970 Washington St. SW, Blacksburg, VA, 24061, USA
| | - Miranda Creasey
- The Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, 970 Washington St. SW, Blacksburg, VA, 24061, USA
| | - Yeonwoo Lebovitz
- The Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, 970 Washington St. SW, Blacksburg, VA, 24061, USA
| | - Michelle H Theus
- The Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, 970 Washington St. SW, Blacksburg, VA, 24061, USA.
| |
Collapse
|
76
|
Ning R, Venkat P, Chopp M, Zacharek A, Yan T, Cui X, Seyfried D, Chen J. D-4F increases microRNA-124a and reduces neuroinflammation in diabetic stroke rats. Oncotarget 2017; 8:95481-95494. [PMID: 29221142 PMCID: PMC5707036 DOI: 10.18632/oncotarget.20751] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 08/15/2017] [Indexed: 01/28/2023] Open
Abstract
D-4F is an apolipoprotein-A1 mimetic peptide that promotes anti-inflammatory effects. MicroRNA-124 is the most abundant brain-specific microRNA and has anti-inflammatory effects. In this study, we investigated the therapeutic efficacy and mechanisms of D-4F treatment of stroke in type one diabetes mellitus (T1DM) rats. Male Wistar rats were induced with T1DM, subjected to embolic middle cerebral artery occlusion and treated with PBS or D-4F (1 mg/kg i.p.) at 2, 24 and 48 hours after stroke (n=8/group). A battery of function tests, brain blood barrier (BBB) integrity, white matter changes and microRNA expression were evaluated in vivo and in vitro. D-4F treatment in T1DM-stroke rats significantly improves functional outcome, decreases BBB leakage, increases tight junction protein expression, decreases white matter damage and inflammatory factor expression, while increasing anti-inflammatory M2 macrophage polarization in the ischemic brain. D-4F significantly increases microRNA-124a expression, and decreases matrix metalloproteinase-9, tumor necrosis factor-α and toll-like receptor-4 gene expression in the ischemic brain, and in primary cortical neuronal and microglial cultures. Inhibition of microRNA-124 in cultured primary cortical neurons and microglia attenuates D-4F induced anti-inflammatory effects and M2 macrophage polarization. D-4F treatment of T1DM-stroke increases microRNA-124 expression, promotes anti-inflammatory effects and M2 macrophage polarization, which may contribute to D-4F-induced improvement in neurological function, and BBB and white matter integrity.
Collapse
Affiliation(s)
- Ruizhuo Ning
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
- Department of Neurology, First Hospital Harbin, Harbin, China
| | - Poornima Venkat
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
- Department of Physics, Oakland University, Rochester, MI, USA
| | - Alex Zacharek
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Tao Yan
- Gerontology Institute, Neurology, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Xu Cui
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Don Seyfried
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Jieli Chen
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
- Gerontology Institute, Neurology, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| |
Collapse
|
77
|
Newton WC, Kim JW, Luo JZQ, Luo L. Stem cell-derived exosomes: a novel vector for tissue repair and diabetic therapy. J Mol Endocrinol 2017; 59:R155-R165. [PMID: 28835418 DOI: 10.1530/jme-17-0080] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 08/23/2017] [Indexed: 12/15/2022]
Abstract
Exosomes are extracellular vesicles (EVs) secreted from a majority of cell types. Exosomes play a role in healthy and pathogenic intercellular interactions via the transfer of proteins, lipids and RNA. The contents and effects of exosomes vary depending on the properties of the originating cell. Exosomes secreted from some cell types, including stem cells, carry biological factors implicated in the protection, regeneration and angiogenesis of damaged tissues. Due to these properties, exosomes have attracted attention as a novel vector for regenerative therapies. Exosomes as a therapeutic tool could have applications for the treatment of many disorders characterized by chronic tissue damage. Exosomes derived from stem cells could be applied to repair or prevent damage from the complications of diabetes mellitus. The immunomodulatory and reparative properties of stem cell-derived exosomes could protect or even restore an early-stage type 1 diabetic patient's original islets from autoimmune destruction. Exosomes could also possibly suppress graft rejection of pancreatic islet transplants. Therefore, it is our recommendation that the treatment of diabetes mellitus using exosome-based therapies be further explored. Development of novel therapies using exosomes is slowed by a limited understanding of their mechanisms. This hurdle must be overcome to pave the way for clinical trials and ultimately the adaptation of exosomes as a therapeutic vector.
Collapse
Affiliation(s)
- William C Newton
- Department of MedicineThe Center of Stem Cell Biology, Roger Williams Hospital, Boston University, School of Medicine, Providence, Rhode Island, USA
| | - Joseph W Kim
- Department of MedicineThe Center of Stem Cell Biology, Roger Williams Hospital, Boston University, School of Medicine, Providence, Rhode Island, USA
| | - John Z Q Luo
- Department of MedicineThe Center of Stem Cell Biology, Roger Williams Hospital, Boston University, School of Medicine, Providence, Rhode Island, USA
- Insure HealthInc., Warwick, Rhode Island, USA
| | - LuGuang Luo
- Department of MedicineThe Center of Stem Cell Biology, Roger Williams Hospital, Boston University, School of Medicine, Providence, Rhode Island, USA
| |
Collapse
|
78
|
Vascular expression of angiopoietin1, α5β1 integrin and tight junction proteins is tightly regulated during vascular remodeling in the post-ischemic brain. Neuroscience 2017; 362:248-256. [DOI: 10.1016/j.neuroscience.2017.08.040] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 06/20/2017] [Accepted: 08/20/2017] [Indexed: 02/05/2023]
|
79
|
Abstract
Neurocardiology is an emerging specialty that addresses the interaction between the brain and the heart, that is, the effects of cardiac injury on the brain and the effects of brain injury on the heart. This review article focuses on cardiac dysfunction in the setting of stroke such as ischemic stroke, brain hemorrhage, and subarachnoid hemorrhage. The majority of post-stroke deaths are attributed to neurological damage, and cardiovascular complications are the second leading cause of post-stroke mortality. Accumulating clinical and experimental evidence suggests a causal relationship between brain damage and heart dysfunction. Thus, it is important to determine whether cardiac dysfunction is triggered by stroke, is an unrelated complication, or is the underlying cause of stroke. Stroke-induced cardiac damage may lead to fatality or potentially lifelong cardiac problems (such as heart failure), or to mild and recoverable damage such as neurogenic stress cardiomyopathy and Takotsubo cardiomyopathy. The role of location and lateralization of brain lesions after stroke in brain-heart interaction; clinical biomarkers and manifestations of cardiac complications; and underlying mechanisms of brain-heart interaction after stroke, such as the hypothalamic-pituitary-adrenal axis; catecholamine surge; sympathetic and parasympathetic regulation; microvesicles; microRNAs; gut microbiome, immunoresponse, and systemic inflammation, are discussed.
Collapse
Affiliation(s)
- Zhili Chen
- From the Gerontology and Neurological Institute, Tianjin Medical University General Hospital, China (Z.C., T.Y., J.C.); Department of Neurology, Henry Ford Hospital, Detroit, MI (P.V., D.S., M.C., J.C.); and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Poornima Venkat
- From the Gerontology and Neurological Institute, Tianjin Medical University General Hospital, China (Z.C., T.Y., J.C.); Department of Neurology, Henry Ford Hospital, Detroit, MI (P.V., D.S., M.C., J.C.); and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Don Seyfried
- From the Gerontology and Neurological Institute, Tianjin Medical University General Hospital, China (Z.C., T.Y., J.C.); Department of Neurology, Henry Ford Hospital, Detroit, MI (P.V., D.S., M.C., J.C.); and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Michael Chopp
- From the Gerontology and Neurological Institute, Tianjin Medical University General Hospital, China (Z.C., T.Y., J.C.); Department of Neurology, Henry Ford Hospital, Detroit, MI (P.V., D.S., M.C., J.C.); and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Tao Yan
- From the Gerontology and Neurological Institute, Tianjin Medical University General Hospital, China (Z.C., T.Y., J.C.); Department of Neurology, Henry Ford Hospital, Detroit, MI (P.V., D.S., M.C., J.C.); and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Jieli Chen
- From the Gerontology and Neurological Institute, Tianjin Medical University General Hospital, China (Z.C., T.Y., J.C.); Department of Neurology, Henry Ford Hospital, Detroit, MI (P.V., D.S., M.C., J.C.); and Department of Physics, Oakland University, Rochester, MI (M.C.).
| |
Collapse
|
80
|
Venkat P, Shen Y, Chopp M, Chen J. Cell-based and pharmacological neurorestorative therapies for ischemic stroke. Neuropharmacology 2017; 134:310-322. [PMID: 28867364 DOI: 10.1016/j.neuropharm.2017.08.036] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 08/22/2017] [Accepted: 08/24/2017] [Indexed: 01/09/2023]
Abstract
Ischemic stroke remains one of most common causes of death and disability worldwide. Stroke triggers a cascade of events leading to rapid neuronal damage and death. Neuroprotective agents that showed promise in preclinical experiments have failed to translate to the clinic. Even after decades of research, tPA remains the only FDA approved drug for stroke treatment. However, tPA is effective when administered 3-4.5 h after stroke onset and the vast majority of stroke patients do not receive tPA therapy. Therefore, there is a pressing need for novel therapies for ischemic stroke. Since stroke induces rapid cell damage and death, neuroprotective strategies that aim to salvage or replace injured brain tissue are challenged by treatment time frames. To overcome the barriers of neuroprotective therapies, there is an increasing focus on neurorestorative therapies for stroke. In this review article, we provide an update on neurorestorative treatments for stroke using cell therapy such as bone marrow derived mesenchymal stromal cells (BMSCs), human umbilical cord blood cells (HUCBCs) and select pharmacological approaches including Minocycline and Candesartan that have been employed in clinical trials. This review article discusses the present understanding of mechanisms of neurorestorative therapies and summarizes ongoing clinical trials. This article is part of the Special Issue entitled 'Cerebral Ischemia'.
Collapse
Affiliation(s)
- Poornima Venkat
- Department of Neurology, Henry Ford Hospital, Detroit, MI, 48202, USA
| | - Yi Shen
- Department of Neurology, Henry Ford Hospital, Detroit, MI, 48202, USA; Gerontology Institute, Department of Neurology, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, 300052, China
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, 48202, USA; Department of Physics, Oakland University, Rochester, MI, 48309, USA
| | - Jieli Chen
- Department of Neurology, Henry Ford Hospital, Detroit, MI, 48202, USA; Gerontology Institute, Department of Neurology, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, 300052, China.
| |
Collapse
|
81
|
Venkat P, Chopp M, Chen J. Blood-Brain Barrier Disruption, Vascular Impairment, and Ischemia/Reperfusion Damage in Diabetic Stroke. J Am Heart Assoc 2017; 6:e005819. [PMID: 28572280 PMCID: PMC5669184 DOI: 10.1161/jaha.117.005819] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Poornima Venkat
- Department of Neurology Research, Henry Ford Hospital, Detroit, MI
| | - Michael Chopp
- Department of Neurology Research, Henry Ford Hospital, Detroit, MI
- Department of Physics, Oakland University, Rochester, MI
| | - Jieli Chen
- Department of Neurology Research, Henry Ford Hospital, Detroit, MI
- Neurological & Gerontology Institute, Neurology, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
82
|
Xiang J, Routhe LJ, Wilkinson DA, Hua Y, Moos T, Xi G, Keep RF. The choroid plexus as a site of damage in hemorrhagic and ischemic stroke and its role in responding to injury. Fluids Barriers CNS 2017; 14:8. [PMID: 28351417 PMCID: PMC5371201 DOI: 10.1186/s12987-017-0056-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 03/08/2017] [Indexed: 12/13/2022] Open
Abstract
While the impact of hemorrhagic and ischemic strokes on the blood–brain barrier has been extensively studied, the impact of these types of stroke on the choroid plexus, site of the blood-CSF barrier, has received much less attention. The purpose of this review is to examine evidence of choroid plexus injury in clinical and preclinical studies of intraventricular hemorrhage, subarachnoid hemorrhage, intracerebral hemorrhage and ischemic stroke. It then discusses evidence that the choroid plexuses are important in the response to brain injury, with potential roles in limiting damage. The overall aim of the review is to highlight deficiencies in our knowledge on the impact of hemorrhagic and ischemic strokes on the choroid plexus, particularly with reference to intraventricular hemorrhage, and to suggest that a greater understanding of the response of the choroid plexus to stroke may open new avenues for brain protection.
Collapse
Affiliation(s)
- Jianming Xiang
- Department of Neurosurgery, University of Michigan, R5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Lisa J Routhe
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - D Andrew Wilkinson
- Department of Neurosurgery, University of Michigan, R5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Ya Hua
- Department of Neurosurgery, University of Michigan, R5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Torben Moos
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, R5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, R5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA. .,Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, USA.
| |
Collapse
|
83
|
Wang Y, Ji X, Leak RK, Chen F, Cao G. Stem cell therapies in age-related neurodegenerative diseases and stroke. Ageing Res Rev 2017; 34:39-50. [PMID: 27876573 PMCID: PMC5250574 DOI: 10.1016/j.arr.2016.11.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Revised: 09/26/2016] [Accepted: 11/04/2016] [Indexed: 02/06/2023]
Abstract
Aging, a complex process associated with various structural, functional and metabolic changes in the brain, is an important risk factor for neurodegenerative diseases and stroke. These diseases share similar neuropathological changes, such as the formation of misfolded proteins, oxidative stress, loss of neurons and synapses, dysfunction of the neurovascular unit (NVU), reduction of self-repair capacity, and motor and/or cognitive deficiencies. In addition to gray matter dysfunction, the plasticity and repair capacity of white matter also decrease with aging and contribute to neurodegenerative diseases. Aging not only renders patients more susceptible to these disorders, but also attenuates their self-repair capabilities. In addition, low drug responsiveness and intolerable side effects are major challenges in the prevention and treatment of senile diseases. Thus, stem cell therapies-characterized by cellular plasticity and the ability to self-renew-may be a promising strategy for aging-related brain disorders. Here, we review the common pathophysiological changes, treatments, and the promises and limitations of stem cell therapies in age-related neurodegenerative diseases and stroke.
Collapse
Affiliation(s)
- Yuan Wang
- Departments of Neurology, Xuanwu Hospital, Capital University of Medicine, Beijing 100053, China
| | - Xunming Ji
- Departments of Neurosurgery, Xuanwu Hospital, Capital University of Medicine, Beijing 100053, China
| | - Rehana K Leak
- Division of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA 15282, United States
| | - Fenghua Chen
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, United States
| | - Guodong Cao
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, United States; Geriatric Research Education and Clinical Centers, VA Pittsburgh Healthcare System, Pittsburgh, PA 15240, United States.
| |
Collapse
|
84
|
Xiang J, Hu J, Shen T, Liu B, Hua F, Zan K, Zu J, Cui G, Ye X. Bone marrow mesenchymal stem cells-conditioned medium enhances vascular remodeling after stroke in type 2 diabetic rats. Neurosci Lett 2017; 644:62-66. [DOI: 10.1016/j.neulet.2017.02.040] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 02/14/2017] [Accepted: 02/15/2017] [Indexed: 10/20/2022]
|
85
|
Kassis H, Shehadah A, Chopp M, Zhang ZG. Epigenetics in Stroke Recovery. Genes (Basel) 2017; 8:genes8030089. [PMID: 28264471 PMCID: PMC5368693 DOI: 10.3390/genes8030089] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 02/02/2017] [Accepted: 02/20/2017] [Indexed: 12/30/2022] Open
Abstract
Abstract: While the death rate from stroke has continually decreased due to interventions in the hyperacute stage of the disease, long-term disability and institutionalization have become common sequelae in the aftermath of stroke. Therefore, identification of new molecular pathways that could be targeted to improve neurological recovery among survivors of stroke is crucial. Epigenetic mechanisms such as post-translational modifications of histone proteins and microRNAs have recently emerged as key regulators of the enhanced plasticity observed during repair processes after stroke. In this review, we highlight the recent advancements in the evolving field of epigenetics in stroke recovery.
Collapse
Affiliation(s)
- Haifa Kassis
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA.
| | - Amjad Shehadah
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA.
| | - Michael Chopp
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA.
- Department of Physics, Oakland University, Rochester, MI 48309, USA.
| | - Zheng Gang Zhang
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA.
| |
Collapse
|
86
|
Venkat P, Chopp M, Chen J. New insights into coupling and uncoupling of cerebral blood flow and metabolism in the brain. Croat Med J 2017; 57:223-8. [PMID: 27374823 PMCID: PMC4937223 DOI: 10.3325/cmj.2016.57.223] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The brain has high metabolic and energy needs and requires continuous cerebral blood flow (CBF), which is facilitated by a tight coupling between neuronal activity, CBF, and metabolism. Upon neuronal activation, there is an increase in energy demand, which is then met by a hemodynamic response that increases CBF. Such regional CBF increase in response to neuronal activation is observed using neuroimaging techniques such as functional magnetic resonance imaging and positron emission tomography. The mechanisms and mediators (eg, nitric oxide, astrocytes, and ion channels) that regulate CBF-metabolism coupling have been extensively studied. The neurovascular unit is a conceptual model encompassing the anatomical and metabolic interactions between the neurons, vascular components, and glial cells in the brain. It is compromised under disease states such as stroke, diabetes, hypertension, dementias, and with aging, all of which trigger a cascade of inflammatory responses that exacerbate brain damage. Hence, tight regulation and maintenance of neurovascular coupling is central for brain homeostasis. This review article also discusses the waste clearance pathways in the brain such as the glymphatic system. The glymphatic system is a functional waste clearance pathway that removes metabolic wastes and neurotoxins from the brain along paravascular channels. Disruption of the glymphatic system burdens the brain with accumulating waste and has been reported in aging as well as several neurological diseases.
Collapse
Affiliation(s)
| | | | - Jieli Chen
- Jieli Chen, Senior Staff Investigator, Henry Ford Hospital, Neurology Research, E&R Building, 3091, Detroit, MI, 48202, USA,
| |
Collapse
|
87
|
Kar S, Bali KK, Baisantry A, Geffers R, Samii A, Bertalanffy H. Genome-Wide Sequencing Reveals MicroRNAs Downregulated in Cerebral Cavernous Malformations. J Mol Neurosci 2017; 61:178-188. [PMID: 28181149 DOI: 10.1007/s12031-017-0880-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 01/05/2017] [Indexed: 12/29/2022]
Abstract
Cerebral cavernous malformations (CCM) are vascular lesions associated with loss-of-function mutations in one of the three genes encoding KRIT1 (CCM1), CCM2, and PDCD10. Recent understanding of the molecular mechanisms that lead to CCM development is limited. The role of microRNAs (miRNAs) has been demonstrated in vascular pathologies resulting in loss of tight junction proteins, increased vascular permeability and endothelial cell dysfunction. Since the relevance of miRNAs in CCM pathophysiology has not been elucidated, the primary aim of the study was to identify the miRNA-mRNA expression network associated with CCM. Using small RNA sequencing, we identified a total of 764 matured miRNAs expressed in CCM patients compared to the healthy brains. The expression of the selected miRNAs was validated by qRT-PCR, and the results were found to be consistent with the sequencing data. Upon application of additional statistical stringency, five miRNAs (let-7b-5p, miR-361-5p, miR-370-3p, miR-181a-2-3p, and miR-95-3p) were prioritized to be top CCM-relevant miRNAs. Further in silico analyses revealed that the prioritized miRNAs have a direct functional relation with mRNAs, such as MIB1, HIF1A, PDCD10, TJP1, OCLN, HES1, MAPK1, VEGFA, EGFL7, NF1, and ENG, which are previously characterized as key regulators of CCM pathology. To date, this is the first study to investigate the role of miRNAs in CCM pathology. By employing cutting edge molecular and in silico analyses on clinical samples, the current study reports global miRNA expression changes in CCM patients and provides a rich source of data set to understand detailed molecular machinery involved in CCM pathophysiology.
Collapse
Affiliation(s)
- Souvik Kar
- International Neuroscience Institute, Rudolf-Pichlmayr-Strasse 4, 30625, Hannover, Germany.
| | - Kiran Kumar Bali
- Pharmacology Institute, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Arpita Baisantry
- Department of Kidney, Liver and Metabolic Diseases, Children's Hospital, Hannover Medical School, Hannover, Germany
| | - Robert Geffers
- Genome Analytics Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Amir Samii
- International Neuroscience Institute, Rudolf-Pichlmayr-Strasse 4, 30625, Hannover, Germany
| | - Helmut Bertalanffy
- International Neuroscience Institute, Rudolf-Pichlmayr-Strasse 4, 30625, Hannover, Germany
| |
Collapse
|
88
|
Liska MG, Crowley MG, Borlongan CV. Regulated and Unregulated Clinical Trials of Stem Cell Therapies for Stroke. Transl Stroke Res 2017; 8:93-103. [PMID: 28127687 DOI: 10.1007/s12975-017-0522-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Accepted: 01/17/2017] [Indexed: 12/17/2022]
Affiliation(s)
- Michael G Liska
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd, Tampa, FL, 33612, USA
| | - Marci G Crowley
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd, Tampa, FL, 33612, USA
| | - Cesar V Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd, Tampa, FL, 33612, USA.
| |
Collapse
|
89
|
Martinez B, Peplow PV. Immunomodulators and microRNAs as neurorestorative therapy for ischemic stroke. Neural Regen Res 2017; 12:865-874. [PMID: 28761412 PMCID: PMC5514854 DOI: 10.4103/1673-5374.208540] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Most of all strokes are ischemic due to occlusion of a vessel, and comprise two main types, thrombotic and embolic. Inflammation and immune response play an important role in the outcome of ischemic stroke. Pharmaceutical and cell-based therapies with immunomodulatory properties could be of benefit in treating ischemic stroke. Possible changes in microRNAs brought about by immunomodulatory treatments may be important. The pharmaceutical studies described in this review have identified several differentially regulated miRNAs associated with disregulation of mRNA targets or the upregulation of several neuroprotective genes, thereby highlighting the potential neuroprotective roles of specific miRNAs such as miR-762, -1892, -200a, -145. MiR-124, -711, -145 are the strongly associated miRNAs predicted to mediate anti-inflammatory pathways and microglia/macrophage M2-like activation phenotype. The cell-based therapy studies reviewed have mainly utilized mesenchymal stem cells or human umbilical cord blood cells and shown to improve functional and neurological outcomes in stroke animals. MiR-145 and miR-133b were implicated in nerve cell remodeling and functional recovery after stroke. Human umbilical cord blood cells decreased proinflammatory factors and promoted M2 macrophage polarization in stroke diabetic animals.
Collapse
Affiliation(s)
- Bridget Martinez
- Department of Molecular and Cellular Biology, University of California, Merced, CA, USA
| | - Philip V Peplow
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| |
Collapse
|
90
|
Bolino A, Piguet F, Alberizzi V, Pellegatta M, Rivellini C, Guerrero-Valero M, Noseda R, Brombin C, Nonis A, D'Adamo P, Taveggia C, Previtali SC. Niacin-mediated Tace activation ameliorates CMT neuropathies with focal hypermyelination. EMBO Mol Med 2016; 8:1438-1454. [PMID: 27799291 PMCID: PMC5167133 DOI: 10.15252/emmm.201606349] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Charcot–Marie–Tooth (CMT) neuropathies are highly heterogeneous disorders caused by mutations in more than 70 genes, with no available treatment. Thus, it is difficult to envisage a single suitable treatment for all pathogenetic mechanisms. Axonal Neuregulin 1 (Nrg1) type III drives Schwann cell myelination and determines myelin thickness by ErbB2/B3‐PI3K–Akt signaling pathway activation. Nrg1 type III is inhibited by the α‐secretase Tace, which negatively regulates PNS myelination. We hypothesized that modulation of Nrg1 levels and/or secretase activity may constitute a unifying treatment strategy for CMT neuropathies with focal hypermyelination as it could restore normal levels of myelination. Here we show that in vivo delivery of Niaspan, a FDA‐approved drug known to enhance TACE activity, efficiently rescues myelination in the Mtmr2−/− mouse, a model of CMT4B1 with myelin outfoldings, and in the Pmp22+/− mouse, which reproduces HNPP (hereditary neuropathy with liability to pressure palsies) with tomacula. Importantly, we also found that Niaspan reduces hypermyelination of Vim (vimentin)−/− mice, characterized by increased Nrg1 type III and Akt activation, thus corroborating the hypothesis that Niaspan treatment downregulates Nrg1 type III signaling.
Collapse
Affiliation(s)
- Alessandra Bolino
- INSPE-Institute of Experimental Neurology, San Raffaele Scientific Institute, Milan, Italy .,Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Françoise Piguet
- INSPE-Institute of Experimental Neurology, San Raffaele Scientific Institute, Milan, Italy.,Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Valeria Alberizzi
- INSPE-Institute of Experimental Neurology, San Raffaele Scientific Institute, Milan, Italy.,Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Marta Pellegatta
- INSPE-Institute of Experimental Neurology, San Raffaele Scientific Institute, Milan, Italy.,Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Cristina Rivellini
- INSPE-Institute of Experimental Neurology, San Raffaele Scientific Institute, Milan, Italy.,Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Marta Guerrero-Valero
- INSPE-Institute of Experimental Neurology, San Raffaele Scientific Institute, Milan, Italy.,Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Roberta Noseda
- INSPE-Institute of Experimental Neurology, San Raffaele Scientific Institute, Milan, Italy.,Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Brombin
- University Centre of Statistics in the Biomedical Sciences (CUSSB), Vita-Salute San Raffaele University, Milan, Italy
| | - Alessandro Nonis
- University Centre of Statistics in the Biomedical Sciences (CUSSB), Vita-Salute San Raffaele University, Milan, Italy
| | - Patrizia D'Adamo
- Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Carla Taveggia
- INSPE-Institute of Experimental Neurology, San Raffaele Scientific Institute, Milan, Italy.,Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Stefano Carlo Previtali
- INSPE-Institute of Experimental Neurology, San Raffaele Scientific Institute, Milan, Italy.,Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy.,Department of Neurology, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
91
|
Napoli E, Borlongan CV. Recent Advances in Stem Cell-Based Therapeutics for Stroke. Transl Stroke Res 2016; 7:452-457. [PMID: 27515852 PMCID: PMC5065756 DOI: 10.1007/s12975-016-0490-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 08/03/2016] [Indexed: 01/27/2023]
Abstract
Regenerative medicine for central nervous system disorders, including stroke, has challenged the non-regenerative capacity of the brain. Among the many treatment strategies tailored towards repairing the injured brain, stem cell-based therapeutics have been demonstrated as safe and effective in animal models of stroke, and are being tested in limited clinical trials. We address here key lab-to-clinic translational research that relate to efficacy, safety, and mechanism of action underlying stem cell therapy. Recognizing the multi-pronged cell death processes associated with stroke that will likely require combination therapies, we next discuss potent drugs and novel technologies directed at improving the functional outcomes of stem cell-based therapeutics. We also examine discrepant transplant regimens between preclinical studies and clinical trials, as well as missing appropriate control arm (i.e., stroke subjects undergoing rehabilitation) on which to directly compare the therapeutic benefits of cell therapy. Finally, the bioethics of cell therapy is presented in order to assess its prevailing social status. With preliminary results now being reported from on-going clinical trials of stem cell therapy for stroke, a careful assessment of the true functional benefits of this novel treatment will further direct the future of regenerative medicine for neurological disorders.
Collapse
Affiliation(s)
- Eleonora Napoli
- Department of Molecular Biosciences, University of California Davis, Davis, CA, USA
| | - Cesar V Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B. Downs Blvd, Tampa, FL, 33612, USA.
| |
Collapse
|
92
|
Pu H, Jiang X, Wei Z, Hong D, Hassan S, Zhang W, Liu J, Meng H, Shi Y, Chen L, Chen J. Repetitive and Prolonged Omega-3 Fatty Acid Treatment After Traumatic Brain Injury Enhances Long-Term Tissue Restoration and Cognitive Recovery. Cell Transplant 2016; 26:555-569. [PMID: 27938482 DOI: 10.3727/096368916x693842] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Traumatic brain injury (TBI) is one of the most disabling clinical conditions that could lead to neurocognitive disorders in survivors. Our group and others previously reported that prophylactic enrichment of dietary omega-3 polyunsaturated fatty acids (n-3 PUFAs) markedly ameliorate cognitive deficits after TBI. However, it remains unclear whether a clinically relevant therapeutic regimen with n-3 PUFAs administered after TBI would still offer significant improvement of long-term cognitive recovery. In the present study, we employed the decline of spatial cognitive function as a main outcome after TBI to investigate the therapeutic efficacy of post-TBI n-3 PUFA treatment and the underlying mechanisms. Mice were subjected to sham operation or controlled cortical impact, followed by random assignment to receive the following four treatments: (1) vehicle control; (2) daily intraperitoneal injections of n-3 PUFAs for 2 weeks, beginning 2 h after TBI; (3) fish oil dietary supplementation throughout the study, beginning 1 day after TBI; or (4) combination of treatments (2) and (3). Spatial cognitive deficits and chronic brain tissue loss, as well as endogenous brain repair processes such as neurogenesis, angiogenesis, and oligodendrogenesis, were evaluated up to 35 days after TBI. The results revealed prominent spatial cognitive deficits and massive tissue loss caused by TBI. Among all mice receiving post-TBI n-3 PUFA treatments, the combined treatment of fish oil dietary supplement and n-3 PUFA injections demonstrated a reproducible beneficial effect in attenuating cognitive deficits although without reducing gross tissue loss. Mechanistically, the combined treatment promoted post-TBI restorative processes in the brain, including generation of immature neurons, microvessels, and oligodendrocytes, each of which was significantly correlated with the improved cognitive recovery. These results indicated that repetitive and prolonged n-3 PUFA treatments after TBI are capable of enhancing brain remodeling and could be developed as a potential therapy to treat TBI victims in the clinic.
Collapse
|
93
|
The cannabinoid beta-caryophyllene (BCP) induces neuritogenesis in PC12 cells by a cannabinoid-receptor-independent mechanism. Chem Biol Interact 2016; 261:86-95. [PMID: 27871898 DOI: 10.1016/j.cbi.2016.11.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 10/22/2016] [Accepted: 11/16/2016] [Indexed: 01/27/2023]
Abstract
Beta-caryophyllene (BCP) is a phytocannabinoid whose neuroprotective activity has been mainly associated with selective activation of cannabinoid-type-2 (CB2) receptors, inhibition of microglial activation and decrease of inflammation. Here, we addressed the potential of BCP to induce neuritogenesis in PC12 cells, a model system for primary neuronal cells that express trkA receptors, respond to NGF and do not express CB2 receptors. We demonstrated that BCP increases the survival and activates the NGF-specific receptor trkA in NGF-deprived PC12 cells, without increasing the expression of NGF itself. The neuritogenic effect of BCP in PC12 cells was abolished by k252a, an inhibitor of the NGF-specific receptor trkA. Accordingly, BCP did not induce neuritogenesis in SH-SY5Y neuroblastoma cells, a neuronal model that does not express trkA receptors and do not respond to NGF. Additionally, we demonstrated that BCP increases the expression of axonal-plasticity-associated proteins (GAP-43, synapsin and synaptophysin) in PC12 cells. It is known that these proteins are up-regulated by NGF in neurons and neuron-like cells, such as PC12 cells. Altogether, these findings suggest that BCP activates trka receptors and induces neuritogenesis by a mechanism independent of NGF or cannabinoid receptors. This is the first study to show such effects of BCP and their beneficial role in neurodegenerative processes should be further investigated.
Collapse
|
94
|
Zhang Q, Chen ZW, Zhao YH, Liu BW, Liu NW, Ke CC, Tan HM. Bone Marrow Stromal Cells Combined With Sodium Ferulate and n-Butylidenephthalide Promote the Effect of Therapeutic Angiogenesis via Advancing Astrocyte-Derived Trophic Factors After Ischemic Stroke. Cell Transplant 2016; 26:229-242. [PMID: 27772541 DOI: 10.3727/096368916x693536] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Being a potential candidate for stroke treatment, bone marrow-derived mesenchymal stem/stromal cells (BM-MSCs) have been demonstrated to be able to enhance angiogenesis and proliferation of reactive astrocytes, which subsequently leads to the amelioration of neurological injury. Increasing evidence further indicates that combining BM-MSCs with certain agents, such as simvastatin, may improve therapeutic effects. Sodium ferulate (SF) and n-butylidenephthalide (BP), two main components of Radix Angelica Sinensis, are proven to be important regulators of stem cells in cell migration, differentiation, and pluripotency maintenance. This study aimed to investigate whether combining BM-MSCs with SF and BP had better therapeutic effect in the treatment of stroke, and the underlying molecular basis for the therapeutic effects was also investigated. The results showed that combination treatment notably reduced neurological injury after stroke and increased the expression of astrocyte-derived vascular endothelial growth factor (VEGF), brain-derived neurotrophic factor (BDNF), and von Willebrand factor-positive vascular density in the ischemic boundary zone as evaluated by immunofluorescence staining. After treatment with BM-MSCs plus SF and BP, astrocytes showed increased expression of VEGF and BDNF by upregulating protein kinase B/mammalian target of rapamycin (AKT/mTOR) expression in an oxygen- and glucose-deprived (OGD) environment. Human umbilical vein endothelial cells (HUVECs) incubated with the conditioned medium (CM) derived from OGD astrocytes treated with BM-MSCs plus SF and BP showed significantly increased migration and tube formation compared with those incubated with the CM derived from OGD astrocytes treated with BM-MSCs alone. These results demonstrate that combination treatment enhances the expression of astrocyte-derived VEGF and BDNF, which contribute to angiogenesis after cerebral ischemia, and the underlying mechanism is associated with activation of the astrocytic AKT/mTOR signaling pathway. Our study provides a potential therapeutic approach for ischemic stroke.
Collapse
|
95
|
Yan T, Venkat P, Chopp M, Zacharek A, Ning R, Roberts C, Zhang Y, Lu M, Chen J. Neurorestorative Responses to Delayed Human Mesenchymal Stromal Cells Treatment of Stroke in Type 2 Diabetic Rats. Stroke 2016; 47:2850-2858. [PMID: 27729575 DOI: 10.1161/strokeaha.116.014686] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 09/14/2016] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND PURPOSE Comorbidity of diabetes mellitus and stroke results in worse functional outcome, poor long-term recovery, and extensive vascular damage. We investigated the neurorestorative effects and mechanisms of stroke treatment with human bone marrow-derived mesenchymal stromal cells (hMSCs) in type 2 diabetes mellitus (T2DM) rats. METHODS Adult male Wistar rats were induced with T2DM, subjected to 2 hours of middle cerebral artery occlusion (MCAo) and treated via tail-vein injection with (1) PBS (n=8) and (2) hMSCs (n=10; 5×106) at 3 days after MCAo. RESULTS In T2DM rats, hMSCs administered at 3 days after MCAo significantly improves neurological function without affecting blood glucose, infarct volume, and incidence of brain hemorrhage in comparison to T2DM-MCAo PBS-treated rats. Delayed hMSC treatment of T2DM stroke significantly improves blood-brain barrier integrity, increases vascular and arterial density and cerebral vascular perfusion, and promotes neuroblast cell migration and white matter remodeling as indicated by increased doublecortin, axon, myelin, and neurofilament density, respectively. Delayed hMSC treatment significantly increases platelet-derived growth factor expression in the ischemic brain, decreases proinflammatory M1 macrophage and increases anti-inflammatory M2 macrophage compared to PBS-treated T2DM-MCAo rats. In vitro data show that hMSCs increase subventricular zone explant cell migration and primary cortical neuron neurite outgrowth, whereas inhibition of platelet-derived growth factor decreases hMSC-induced subventricular zone cell migration and axonal outgrowth. CONCLUSIONS In T2DM stroke rats, delayed hMSC treatment significantly improves neurological functional outcome and increases neurorestorative effects and M2 macrophage polarization. Increasing brain platelet-derived growth factor expression may contribute to hMSC-induced neurorestoration.
Collapse
Affiliation(s)
- Tao Yan
- From the Tianjin Neurological and Gerontology Institute, Department of Neurology of Tianjin Medical University General Hospital, China (T.Y., J.C.); Department of Neurology (T.Y., P.V., M.C., A.Z., R.N., C.R., Y.Z., J.C.) and Department of Biostatistics and Research Epidemiology (M.L.), Henry Ford Hospital, Detroit, MI; and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Poornima Venkat
- From the Tianjin Neurological and Gerontology Institute, Department of Neurology of Tianjin Medical University General Hospital, China (T.Y., J.C.); Department of Neurology (T.Y., P.V., M.C., A.Z., R.N., C.R., Y.Z., J.C.) and Department of Biostatistics and Research Epidemiology (M.L.), Henry Ford Hospital, Detroit, MI; and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Michael Chopp
- From the Tianjin Neurological and Gerontology Institute, Department of Neurology of Tianjin Medical University General Hospital, China (T.Y., J.C.); Department of Neurology (T.Y., P.V., M.C., A.Z., R.N., C.R., Y.Z., J.C.) and Department of Biostatistics and Research Epidemiology (M.L.), Henry Ford Hospital, Detroit, MI; and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Alex Zacharek
- From the Tianjin Neurological and Gerontology Institute, Department of Neurology of Tianjin Medical University General Hospital, China (T.Y., J.C.); Department of Neurology (T.Y., P.V., M.C., A.Z., R.N., C.R., Y.Z., J.C.) and Department of Biostatistics and Research Epidemiology (M.L.), Henry Ford Hospital, Detroit, MI; and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Ruizhuo Ning
- From the Tianjin Neurological and Gerontology Institute, Department of Neurology of Tianjin Medical University General Hospital, China (T.Y., J.C.); Department of Neurology (T.Y., P.V., M.C., A.Z., R.N., C.R., Y.Z., J.C.) and Department of Biostatistics and Research Epidemiology (M.L.), Henry Ford Hospital, Detroit, MI; and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Cynthia Roberts
- From the Tianjin Neurological and Gerontology Institute, Department of Neurology of Tianjin Medical University General Hospital, China (T.Y., J.C.); Department of Neurology (T.Y., P.V., M.C., A.Z., R.N., C.R., Y.Z., J.C.) and Department of Biostatistics and Research Epidemiology (M.L.), Henry Ford Hospital, Detroit, MI; and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Yi Zhang
- From the Tianjin Neurological and Gerontology Institute, Department of Neurology of Tianjin Medical University General Hospital, China (T.Y., J.C.); Department of Neurology (T.Y., P.V., M.C., A.Z., R.N., C.R., Y.Z., J.C.) and Department of Biostatistics and Research Epidemiology (M.L.), Henry Ford Hospital, Detroit, MI; and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Mei Lu
- From the Tianjin Neurological and Gerontology Institute, Department of Neurology of Tianjin Medical University General Hospital, China (T.Y., J.C.); Department of Neurology (T.Y., P.V., M.C., A.Z., R.N., C.R., Y.Z., J.C.) and Department of Biostatistics and Research Epidemiology (M.L.), Henry Ford Hospital, Detroit, MI; and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Jieli Chen
- From the Tianjin Neurological and Gerontology Institute, Department of Neurology of Tianjin Medical University General Hospital, China (T.Y., J.C.); Department of Neurology (T.Y., P.V., M.C., A.Z., R.N., C.R., Y.Z., J.C.) and Department of Biostatistics and Research Epidemiology (M.L.), Henry Ford Hospital, Detroit, MI; and Department of Physics, Oakland University, Rochester, MI (M.C.).
| |
Collapse
|
96
|
Liu X, Hou L, Huang W, Gao Y, Lv X, Tang J. The Mechanism of Long Non-coding RNA MEG3 for Neurons Apoptosis Caused by Hypoxia: Mediated by miR-181b-12/15-LOX Signaling Pathway. Front Cell Neurosci 2016; 10:201. [PMID: 27642276 PMCID: PMC5009716 DOI: 10.3389/fncel.2016.00201] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 08/03/2016] [Indexed: 02/01/2023] Open
Abstract
Objective: lncRNAs are recently thought to play a significant role in cellular homeostasis during pathological process of diseases by competing inhibiting miRNA function. The aim of present study was to assess the function of long non-coding RNA (lncRNA) MEG3 and its functional interaction with microRNA-181b in cerebral ischemic infarct of mice and hypoxia-induced neurons apoptosis. Methods: To address this question, we performed the experiments with in vivo middle cerebral artery occlusion (MCAO) mice model and in vitro oxygen-glucose deprivation (OGD)-cultured neuronal HT22 cell line. Relative expression of MEG3, miR-181b, and 12/15-LOX (lipoxygenase) mRNA was determined using quantitative RT-PCR. Western blot was used to evaluate 12/15-LOX protein expression. TUNEL assay was performed to assess cell apoptosis. Results: In both MCAO mice and OGD-cultured HT22 cell, ischemia, or hypoxia treatment results in a time-dependent increase in MEG3 and 12/15-LOX expression and decrease in miR-181b expression. Knockdown of MEG3 contributes to attenuation of hypoxia-induced apoptosis of HT22 cell. Also, expression level of MEG3 negatively correlated with miR-181b expression and positively correlated with 12/15-LOX expression. In contrary to MEG3, miR-181b overexpression attenuated hypoxia-induced HT22 cell apoptosis, as well as suppressed hypoxia-induced increase in 12/15-LOX expression. By luciferase reporter assay, we concluded that miR-181b directly binds to 12/15-LOX 3′-UTR, thereby negatively regulates 12/15-LOX expression. Conclusion: Our data suggested that long non-coding RNA MEG3 functions as a competing endogenous RNA for miR-181b to regulate 12/15-LOX expression in middle cerebral artery occlusion-induced ischemic infarct of brain nerve cells.
Collapse
Affiliation(s)
- Xiaomin Liu
- Department of Neurology, Qianfoshan Hospital, Shandong University Jinan, China
| | - Lijing Hou
- Department of Pharmacological Laboratory, Shandong Academy of Chinese Medicine Jinan, China
| | - Weiwei Huang
- Department of Neurology, Qianfoshan Hospital, Shandong University Jinan, China
| | - Yuan Gao
- Department of Neurology, Qianfoshan Hospital, Shandong University Jinan, China
| | - Xin Lv
- Department of Neurology, Qianfoshan Hospital, Shandong University Jinan, China
| | - Jiyou Tang
- Department of Neurology, Qianfoshan Hospital, Shandong University Jinan, China
| |
Collapse
|
97
|
Human endothelial progenitor cells rescue cortical neurons from oxygen-glucose deprivation induced death. Neurosci Lett 2016; 631:50-55. [PMID: 27521752 DOI: 10.1016/j.neulet.2016.08.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 07/12/2016] [Accepted: 08/09/2016] [Indexed: 12/22/2022]
Abstract
BACKGROUND AND AIM Cerebral ischemia is characterized by both acute and delayed neuronal injuries. Neuro-protection is a major issue that should be properly addressed from a pharmacological point of view, and cell-based treatment approaches are of interest due to their potential pleiotropic effects. Endothelial progenitor cells have the advantage of being mobilized from the bone marrow into the circulation, but have been less studied than other stem cells, such as mesenchymal stem cells. Therefore, the comparison between human endothelial progenitor cells (hEPC) and human mesenchymal progenitor cells (hMSC) in terms of efficacy in rescuing neurons from cell death after transitory ischemia is the aim of the current study, in the effort to address further directions. MATERIALS AND METHODS In vitro model of oxygen-glucose deprivation (OGD) on a primary culture of rodent cortical neurons was set up with different durations of exposure: 1, 2 and 3hrs with assessment of neuron survival. The 2hrs OGD was chosen for the subsequent experiments. After 2hrs OGD neurons were either placed in indirect co-culture with hMSC or hEPC or cultured in hMSC or hEPC conditioned medium and cell viability was evaluated by MTT assay. RESULTS At day 2 after 2hrs OGD exposure, mean neuronal survival was 47.9±24.2%. In contrast, after treatment with hEPC and hMSC indirect co-culture was 74.1±27.3%; and 69.4±18.8%, respectively. In contrast, treatment with conditioned medium did not provide any advantage in terms of survival to OGD neurons CONCLUSION The study shows the efficacy of hEPC in indirect co-culture to rescue neurons from cell death after OGD, comparable to that of hMSC. hEPC deserve further studies given their potential interest for ischemia.
Collapse
|
98
|
Jhelum P, Reddy RG, Kumar A, Chakravarty S. Natural product based novel small molecules with promising neurotrophic, neurogenic and anti-neuroinflammatory actions can be developed as stroke therapeutics. Neural Regen Res 2016; 11:916-7. [PMID: 27482216 PMCID: PMC4962585 DOI: 10.4103/1673-5374.184486] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Priya Jhelum
- Chemical Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
| | - R Gajendra Reddy
- Chemical Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
| | - Arvind Kumar
- CSIR- Centre for Cellular and Molecular Biology, Hyderabad, India
| | - Sumana Chakravarty
- Chemical Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
| |
Collapse
|
99
|
Zhang Q, Zhao Y, Xu Y, Chen Z, Liu N, Ke C, Liu B, Wu W. Sodium ferulate and n-butylidenephthalate combined with bone marrow stromal cells (BMSCs) improve the therapeutic effects of angiogenesis and neurogenesis after rat focal cerebral ischemia. J Transl Med 2016; 14:223. [PMID: 27465579 PMCID: PMC4963939 DOI: 10.1186/s12967-016-0979-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Accepted: 07/13/2016] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Studies have indicated that bone marrow stromal cell (BMSC) administration is a promising approach for stroke treatment. For our study, we chose sodium ferulate (SF) and n-butylidenephthalide (BP) combined with BMSC, and observed if the combination treatment possessed more significant effects on angiogenesis and neurogenesis post-stroke. METHODS We established rat permanent middle cerebral artery occlusion (MCAo) model and evaluated ischemic volumes of MCAo, BMSC, SF + BP, Simvastatin + BMSC and SF + BP + BMSC groups with TTC staining on the 7th day after ischemia. Immunofluorescence staining of vascular endothelial growth factor (VEGF) and brain derived neurotrophic factor (BDNF), as well as immunohistochemistry staining of von Willebrand factor (vWF) and neuronal class III β-tubulin (Tuj1) were performed in ischemic boundary zone (IBZ), furthermore, to understand the mechanism, western blot was used to investigate AKT/mammalian target of rapamycin (mTOR) signal pathway in ischemic cortex. We also tested BMSC derived-VEGF and BDNF expressions by western blot assay in vitro. RESULTS SF + BP + BMSC group obviously decreased infarction zone, and elevated the expression of VEGF and the density and perimeter of vWF-vessels as same as Simvastatin + BMSC administration; moreover, its effects on BDNF and Tuj1 expressions were superior to Simvastatin + BMSC treatment in IBZ. Meanwhile, it showed that SF and BP combined with BMSC treatment notably up-regulated AKT/mTOR signal pathway compared with SF + BP group and BMSC alone post-stroke. Western blot results showed that SF and BP treatment could promote BMSCs to synthesize VEGF and BDNF in vitro. CONCLUSIONS We firstly demonstrate that SF and BP combined with BMSC can significantly improve angiogenesis and neurogenesis in IBZ following stroke. The therapeutic effects are associated with the enhancement of VEGF and BDNF expressions via activation of AKT/mTOR signal pathway. Furthermore, triggering BMSC paracrine function of SF and BP might contribute to amplifying the synergic effects of the combination treatment.
Collapse
Affiliation(s)
- Qian Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao, People's Republic of China
| | - Yonghua Zhao
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao, People's Republic of China.
| | - Youhua Xu
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao, People's Republic of China
| | - Zhenwei Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao, People's Republic of China
| | - Naiwei Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao, People's Republic of China
| | - Chienchih Ke
- Biomedical Imaging Research Center, National Yang Ming University, Taipei, Taiwan
| | - Bowen Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao, People's Republic of China
| | - Weikang Wu
- Department of pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People's Republic of China
| |
Collapse
|
100
|
Cui C, Ye X, Chopp M, Venkat P, Zacharek A, Yan T, Ning R, Yu P, Cui G, Chen J. miR-145 Regulates Diabetes-Bone Marrow Stromal Cell-Induced Neurorestorative Effects in Diabetes Stroke Rats. Stem Cells Transl Med 2016; 5:1656-1667. [PMID: 27460851 PMCID: PMC5189645 DOI: 10.5966/sctm.2015-0349] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 05/13/2016] [Indexed: 01/08/2023] Open
Abstract
In rats with type 1 diabetes mellitus (T1DM) subject to stroke, the therapeutic effects and underlying mechanisms of action of bone-marrow stromal cells (BMSCs) derived from T1DM rats (DM-BMSCs) and BMSCs derived from normal rats (Nor-BMSCs) were compared. In vitro and in vivo, DM-BMSCs exhibited decreased miR-145 expression. In T1DM rats, DM-BMSC treatment significantly improved functional outcome and increased vascular and white matter remodeling. However, overexpression of miR-145 in DM-BMSCs attenuates DM-BMSC-induced neurorestorative effects in T1DM stroke rats. In rats with type 1 diabetes (T1DM), the therapeutic effects and underlying mechanisms of action of stroke treatment were compared between bone-marrow stromal cells (BMSCs) derived from T1DM rats (DM-BMSCs) and BMSCs derived from normal rats (Nor-BMSCs). The novel role of microRNA-145 (miR-145) in mediating DM-BMSC treatment-induced benefits was also investigated. T1DM rats (n = 8 per group) underwent 2 hours of middle cerebral artery occlusion (MCAo) and were treated 24 hours later with the one of the following (5 × 106 cells administered i.v.): (a) phosphate-buffered saline (PBS); (b) Nor-BMSCs; (c) DM-BMSCs; (d) DM-BMSCs with miR-145 overexpression (miR-145+/+DM-BMSCs); or (e) Nor-BMSCs with miR-145 knockdown. Evaluation of functional outcome, vascular and white-matter remodeling and microRNA expression was made, and in vitro studies were performed. In vitro, DM-BMSCs exhibited decreased miR-145 expression and increased survival compared with Nor-BMSCs. Capillary tube formation and axonal outgrowth in cultured primary cortical neurons were significantly increased by DM-BMSC-conditioned medium compared with Nor-BMSCs, and significantly decreased by miR-145+/+DM-BMSC-conditioned medium compared with DM-BMSCs. In T1DM rats in which stroke had been induced (T1DM stroke rats), DM-BMSC treatment significantly improved functional outcome, increased vascular and white matter remodeling, decreased serum miR-145 expression, and increased expression of the miR-145 target genes adenosine triphosphate-binding cassette transporter 1 (ABCA1) and insulin-like growth factor 1 receptor (IGFR1), compared with Nor-BMSCs or PBS treatment. However, miR-145+/+DM-BMSCs significantly increased serum miR-145 expression and decreased brain ABCA1 and IGFR1 expression, as well as attenuated DM-BMSC-induced neurorestorative effects in T1DM-MCAo rats. DM-BMSCs exhibited decreased miR-145 expression. In T1DM-MCAo rats, DM-BMSC treatment improved functional outcome and promoted neurorestorative effects. The miR-145/ABCA1/IGFR1 pathway may contribute to the enhanced DM-BMSCs’ functional and neurorestorative effects in T1DM stroke rats. Significance In rats with type 1 diabetes (T1DM), the therapeutic effects and underlying mechanisms of action of stroke treatment were compared between bone-marrow stromal cells (BMSCs) derived from T1DM rats (DM-BMSCs) and BMSCs derived from normal rats (Nor-BMSCs). In vitro, DM-BMSCs and derived exosomes decreased miR-145 expression and increased DM-BMSC survival, capillary tube formation, and axonal outgrowth, compared with Nor-BMSCs; these effects were decreased by DM-BMSCs in which miR-145 was overexpressed. In vivo, compared with Nor-BMSC or phosphate-buffered saline treatment, DM-BMSC treatment improved functional outcome and vascular and white matter remodeling, decreased serum miR-145 expression, and increased expression of the miR-145 target genes ABCA1 and IGFR1. microRNA-145 mediated the benefits induced by DM-BMSC treatment.
Collapse
Affiliation(s)
- Chengcheng Cui
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, People's Republic of China
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Xinchun Ye
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, People's Republic of China
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
- Department of Physics, Oakland University, Rochester, Michigan, USA
| | - Poornima Venkat
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
- Department of Physics, Oakland University, Rochester, Michigan, USA
| | - Alex Zacharek
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Tao Yan
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Ruizhou Ning
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Peng Yu
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Guiyun Cui
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, People's Republic of China
| | - Jieli Chen
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
- Department of Geriatrics, Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| |
Collapse
|