1
|
Larionov A, Hammer CM, Fiedler K, Filgueira L. Dynamics of Endothelial Cell Diversity and Plasticity in Health and Disease. Cells 2024; 13:1276. [PMID: 39120307 PMCID: PMC11312403 DOI: 10.3390/cells13151276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/19/2024] [Accepted: 07/19/2024] [Indexed: 08/10/2024] Open
Abstract
Endothelial cells (ECs) are vital structural units of the cardiovascular system possessing two principal distinctive properties: heterogeneity and plasticity. Endothelial heterogeneity is defined by differences in tissue-specific endothelial phenotypes and their high predisposition to modification along the length of the vascular bed. This aspect of heterogeneity is closely associated with plasticity, the ability of ECs to adapt to environmental cues through the mobilization of genetic, molecular, and structural alterations. The specific endothelial cytoarchitectonics facilitate a quick structural cell reorganization and, furthermore, easy adaptation to the extrinsic and intrinsic environmental stimuli, known as the epigenetic landscape. ECs, as universally distributed and ubiquitous cells of the human body, play a role that extends far beyond their structural function in the cardiovascular system. They play a crucial role in terms of barrier function, cell-to-cell communication, and a myriad of physiological and pathologic processes. These include development, ontogenesis, disease initiation, and progression, as well as growth, regeneration, and repair. Despite substantial progress in the understanding of endothelial cell biology, the role of ECs in healthy conditions and pathologies remains a fascinating area of exploration. This review aims to summarize knowledge and concepts in endothelial biology. It focuses on the development and functional characteristics of endothelial cells in health and pathological conditions, with a particular emphasis on endothelial phenotypic and functional heterogeneity.
Collapse
Affiliation(s)
- Alexey Larionov
- Faculty of Science and Medicine, Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700 Fribourg, Switzerland; (C.M.H.); (L.F.)
| | - Christian Manfred Hammer
- Faculty of Science and Medicine, Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700 Fribourg, Switzerland; (C.M.H.); (L.F.)
| | - Klaus Fiedler
- Independent Researcher, CH-1700 Fribourg, Switzerland;
| | - Luis Filgueira
- Faculty of Science and Medicine, Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700 Fribourg, Switzerland; (C.M.H.); (L.F.)
| |
Collapse
|
2
|
Hu B, Pei J, Wan C, Liu S, Xu Z, Zou Y, Li Z, Tang Z. Mechanisms of Postischemic Stroke Angiogenesis: A Multifaceted Approach. J Inflamm Res 2024; 17:4625-4646. [PMID: 39045531 PMCID: PMC11264385 DOI: 10.2147/jir.s461427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 06/26/2024] [Indexed: 07/25/2024] Open
Abstract
Ischemic stroke constitutes a significant global health care challenge, and a comprehensive understanding of its recovery mechanisms is imperative for the development of innovative therapeutic strategies. Angiogenesis, a pivotal element of ischemic tissue repair, facilitates the restoration of blood flow to damaged regions, thereby promoting neuronal regeneration and functional recovery. Nevertheless, the mechanisms underlying postischemic stroke angiogenesis remain incompletely elucidated. This review meticulously examines the constituents of the neurovascular unit, ion channels, molecular mediators, and signaling pathways implicated in angiogenesis following stroke. Furthermore, it delves into prospective therapeutic strategies informed by these factors. Our objective is to provide detailed and exhaustive information on the intricate mechanisms governing postischemic stroke angiogenesis, thus providing a robust scientific foundation for the advancement of novel neurorepair therapies.
Collapse
Affiliation(s)
- Bin Hu
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Jingchun Pei
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Cheng Wan
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
- Department of Medical Imaging, The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Shuangshuang Liu
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Zhe Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, People’s Republic of China
- School of Basic Medical Sciences, Qujing Medical College, Qujing, People’s Republic of China
| | - Yongwei Zou
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Zhigao Li
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Zhiwei Tang
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| |
Collapse
|
3
|
Shen Z, Xiang M, Chen C, Ding F, Wang Y, Shang C, Xin L, Zhang Y, Cui X. Glutamate excitotoxicity: Potential therapeutic target for ischemic stroke. Biomed Pharmacother 2022; 151:113125. [PMID: 35609367 DOI: 10.1016/j.biopha.2022.113125] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/01/2022] [Accepted: 05/13/2022] [Indexed: 11/29/2022] Open
Abstract
Glutamate-mediated excitotoxicity is an important mechanism leading to post ischemic stroke damage. After acute stroke, the sudden reduction in cerebral blood flow is most initially followed by ion transport protein dysfunction and disruption of ion homeostasis, which in turn leads to impaired glutamate release, reuptake, and excessive N-methyl-D-aspartate receptor (NMDAR) activation, promoting neuronal death. Despite extensive evidence from preclinical studies suggesting that excessive NMDAR stimulation during ischemic stroke is a central step in post-stroke damage, NMDAR blockers have failed to translate into clinical stroke treatment. Current treatment options for stroke are very limited, and there is therefore a great need to develop new targets for neuroprotective therapeutic agents in ischemic stroke to extend the therapeutic time window. In this review, we highlight recent findings on glutamate release, reuptake mechanisms, NMDAR and its downstream cellular signaling pathways in post-ischemic stroke damage, and review the pathological changes in each link to help develop viable new therapeutic targets. We then also summarize potential neuroprotective drugs and therapeutic approaches for these new targets in the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Zihuan Shen
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Clinical Medical School, Beijing University of Traditional Chinese Medicine, Beijing 100029, China
| | - Mi Xiang
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Chen Chen
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Fan Ding
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Clinical Medical School, Beijing University of Traditional Chinese Medicine, Beijing 100029, China
| | - Yuling Wang
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Clinical Medical School, Beijing University of Traditional Chinese Medicine, Beijing 100029, China
| | - Chang Shang
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Clinical Medical School, Beijing University of Traditional Chinese Medicine, Beijing 100029, China
| | - Laiyun Xin
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Yang Zhang
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Xiangning Cui
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
4
|
Abstract
Recent evidence shows that when ischemic stroke (IS) occurs, the BBB would be destructed, thereby promoting the immune cells to migrate into the brain, suggesting that the immune responses can play a vital role in the pathology of IS. As an essential subpopulation of immunosuppressive T cells, regulatory T (Treg) cells are involved in maintaining immune homeostasis and suppressing immune responses in the pathophysiological conditions of IS. During the past decades, the regulatory role of Treg cells has attracted the interest of numerous researchers. However, whether they are beneficial or detrimental to the outcomes of IS remains controversial. Moreover, Treg cells exert distinctive effects in the different stages of IS. Therefore, it is urgent to elucidate how Treg cells modulate the immune responses induced by IS. In this review, we describe how Treg cells fluctuate and play a role in the regulation of immune responses after IS in both experimental animals and humans, and summarize their biological functions and mechanisms in both CNS and periphery. We also discuss how Treg cells participate in poststroke inflammation and immunodepression and the potential of Treg cells as a novel therapeutic approach.
Collapse
|
5
|
Chen X, Sun W, Zhong P, Wu D. Colony-Stimulating Factors on Mobilizing CD34 + Cells and Improving Neurological Functions in Patients With Stroke: A Meta-Analysis and a Systematic Review. Front Pharmacol 2021; 12:704509. [PMID: 34366857 PMCID: PMC8339259 DOI: 10.3389/fphar.2021.704509] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 06/07/2021] [Indexed: 11/13/2022] Open
Abstract
Background and Purpose: CSF therapy is considered a promising therapeutic approach for stroke. We performed a meta-analysis to explore the safety and efficacy of CSF in published clinical stroke studies. Methods: We searched articles online and manually. Two reviewers selected studies independently, selecting data based on study quality, characteristics of intervention (administration time, observation time, type, dose, and injection approach of CSF), and the baseline characteristics of patients (age, sex, hypertension, diabetes, smoker, and lipids) were extracted. Main prognosis outcomes were measured as all-cause death in severe adverse events (SAE) and recurrent stroke in SAE. Secondary outcomes were measured as CD34+ cell counts in periphery blood at day 5, National Institutes of Health Stroke Scale (NIHSS), and Barthel index (BI), Side effects of CSF were taken as the indicator of safety. STATA13 software was used to perform the meta-analysis.Keywords: Stroke, Colony-stimulating factor, Meta-analysis, therapy, Neurological Diseases Results: This meta-analysis involved 485 patients from eight studies. Among them, 475 patients from seven studies were gauged SAE (all-cause death), 393 patients from six studies were checked SAE (recurrent stroke); 137 patients from three studies underwent CD34+ measurement, 389 patients from six studies were tested NIHSS and 307 patients from five studies accessed BI. Compared with the control group, both all-causes death (RR= 1.73, 95%CI= (0.61, 4.92), P=0.735, I2=0.0%) and recurrent stroke (RR= 0.43, 95%CI= (0.14, 1.32), P=0.214, I2=33.1%) present no statistical differences, indicating that the application of CSF does not statistically alter the prognosis of patients with stroke. The application of CSF effectively enhanced CD34+ cell counts in periphery blood at day 5 (SMD= 1.23, 95%CI= (0.54, 1.92), P=0.04, I2=69.0%) but did not statistically impact NIHSS (SMD= -0.40, 95%CI= (-0.93, 0.13), P ≤ 0.001, I2=79.7%) or BI (SMD= 0.04, 95%CI= (-0.38, 0.46), P=0.068, I2=54.3%). Conclusion: Our study consolidates the security of CSF administration for its exerting no effect on detrimental outcomes. It has proven to be effective in elevating CD34+ cell counts in periphery blood at day 5, indicating CSF may participate in stroke recovery, but its efficacy in stroke recovery remains detected.
Collapse
Affiliation(s)
- Xiuqi Chen
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Wenbo Sun
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Ping Zhong
- Department of Neurology, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, China
| | - Danhong Wu
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| |
Collapse
|
6
|
Saad MAEL, Fahmy MIM, Al-Shorbagy M, Assaf N, Hegazy AAEA, El-Yamany MF. Nateglinide Exerts Neuroprotective Effects via Downregulation of HIF-1α/TIM-3 Inflammatory Pathway and Promotion of Caveolin-1 Expression in the Rat's Hippocampus Subjected to Focal Cerebral Ischemia/Reperfusion Injury. Inflammation 2021; 43:401-416. [PMID: 31863220 DOI: 10.1007/s10753-019-01154-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ischemic stroke is a major cause of death and motor disabilities all over the world. It is a muti-factorial disorder associated with inflammatory, apoptotic, and oxidative responses. Nateglinide (NAT), an insulinotropic agent used for the treatment of type 2 diabetes mellitus, recently showed potential anti-inflammatory and anti-apoptotic effects. The aim of our study was to elucidate the unique neuroprotective role of NAT in the middle cerebral artery occlusion (MCAO)-induced stroke in rats. Fifty-six male rats were divided to 4 groups (n = 14 in each group): the sham-operated group, sham receiving NAT (50 mg/kg/day, p.o) group, ischemia/reperfusion (IR) group, and IR receiving NAT group (50 mg/kg/day, p.o). MCAO caused potent deficits in motor and behavioral functions of the rats. Significant increase in inflammatory and apoptotic biomarkers has been observed in rats' hippocampi. Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) pathway was significantly stimulated causing activation of series inflammatory biomarkers ending up neuro-inflammatory milieu. Pretreatment with NAT preserved rats' normal behavioral and motor functions. Moreover, NAT opposed the expression of hypoxia-inducible factor-1α (HIF-1α) resulting in downregulation of more inflammatory mediators namely, NF-κB, tumor necrosis factor-β (TNF-β), and the anti-survival gene PMAIP-1. NAT stimulated caveolin-1 (Cav-1) which prevented expression of oxidative biomarkers, nitric oxide (NO), and myeloperoxidase (MPO) and hamper the activation of apoptotic biomarker caspase-3. In conclusion, our work postulated that NAT exhibited its neuroprotective effects in rats with ischemic stroke via attenuation of different unique oxidative, apoptotic, and inflammatory pathways.
Collapse
Affiliation(s)
- Muhammad Abd El-Latif Saad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Giza, Egypt.,School of Pharmacy, NewGiza University, Giza, Egypt
| | - Mohamed Ibrahim Mohamed Fahmy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy and Drug Technology, Heliopolis University for Sustainable Development, Cairo, Egypt.
| | - Muhammad Al-Shorbagy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Giza, Egypt.,School of Pharmacy, NewGiza University, Giza, Egypt
| | - Naglaa Assaf
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Misr University for Science and Technology (MUST), Giza, Egypt
| | | | | |
Collapse
|
7
|
Lee JY, Castelli V, Bonsack B, García-Sánchez J, Kingsbury C, Nguyen H, Tajiri N, Borlongan CV. Eyeballing stroke: Blood flow alterations in the eye and visual impairments following transient middle cerebral artery occlusion in adult rats. Cell Transplant 2021; 29:963689720905805. [PMID: 32098493 PMCID: PMC7444237 DOI: 10.1177/0963689720905805] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Middle cerebral artery occlusion in rodents remains a widely used model
of ischemic stroke. Recently, we reported the occurrence of retinal
ischemia in animals subjected to middle cerebral artery occlusion,
owing in part to the circulatory juxtaposition of the ophthalmic
artery to the middle cerebral artery. In this study, we examined the
eye hemodynamics and visual deficits in middle cerebral artery
occlusion-induced stroke rats. The brain and eye were evaluated by
laser Doppler at baseline (prior to middle cerebral artery occlusion),
during and after middle cerebral artery occlusion. Retinal
function-relevant behavioral and histological outcomes were performed
at 3 and 14 days post-middle cerebral artery occlusion. Laser Doppler
revealed a typical reduction of at least 80% in the ipsilateral
frontoparietal cortical area of the brain during middle cerebral
artery occlusion compared to baseline, which returned to near-baseline
levels during reperfusion. Retinal perfusion defects closely
paralleled the timing of cerebral blood flow alterations in the acute
stages of middle cerebral artery occlusion in adult rats,
characterized by a significant blood flow defect in the ipsilateral
eye with at least 90% reduction during middle cerebral artery
occlusion compared to baseline, which was restored to near-baseline
levels during reperfusion. Moreover, retinal ganglion cell density and
optic nerve depth were significantly decreased in the ipsilateral eye.
In addition, the stroke rats displayed eye closure. Behavioral
performance in a light stimulus-mediated avoidance test was
significantly impaired in middle cerebral artery occlusion rats
compared to control animals. In view of visual deficits in stroke
patients, closely monitoring of brain and retinal perfusion via laser
Doppler measurements and examination of visual impairments may
facilitate the diagnosis and the treatment of stroke, including
retinal ischemia.
Collapse
Affiliation(s)
- Jea-Young Lee
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, USA
| | - Vanessa Castelli
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, USA
| | - Brooke Bonsack
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, USA
| | - Julián García-Sánchez
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, USA
| | - Chase Kingsbury
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, USA
| | - Hung Nguyen
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, USA
| | - Naoki Tajiri
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, 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, USA
| |
Collapse
|
8
|
Alwjwaj M, Kadir RRA, Bayraktutan U. The secretome of endothelial progenitor cells: a potential therapeutic strategy for ischemic stroke. Neural Regen Res 2021; 16:1483-1489. [PMID: 33433461 PMCID: PMC8323700 DOI: 10.4103/1673-5374.303012] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Ischemic stroke continues to be a leading cause of mortality and morbidity in the world. Despite recent advances in the field of stroke medicine, thrombolysis with recombinant tissue plasminogen activator remains as the only pharmacological therapy for stroke patients. However, due to short therapeutic window (4.5 hours of stroke onset) and increased risk of hemorrhage beyond this point, each year globally less than 1% of stroke patients receive this therapy which necessitate the discovery of safe and efficacious therapeutics that can be used beyond the acute phase of stroke. Accumulating evidence indicates that endothelial progenitor cells (EPCs), equipped with an inherent capacity to migrate, proliferate and differentiate, may be one such therapeutics. However, the limited availability of EPCs in peripheral blood and early senescence of few isolated cells in culture conditions adversely affect their application as effective therapeutics. Given that much of the EPC-mediated reparative effects on neurovasculature is realized by a wide range of biologically active substances released by these cells, it is possible that EPC-secretome may serve as an important therapeutic after an ischemic stroke. In light of this assumption, this review paper firstly discusses the main constituents of EPC-secretome that may exert the beneficial effects of EPCs on neurovasculature, and then reviews the currently scant literature that focuses on its therapeutic capacity.
Collapse
Affiliation(s)
- Mansour Alwjwaj
- Stroke, Division of Clinical Neuroscience, University of Nottingham, Clinical Sciences Building, City Hospital, Hucknall Road, Nottingham, NG5 1PB, UK
| | - Rais Reskiawan A Kadir
- Stroke, Division of Clinical Neuroscience, University of Nottingham, Clinical Sciences Building, City Hospital, Hucknall Road, Nottingham, NG5 1PB, UK
| | - Ulvi Bayraktutan
- Stroke, Division of Clinical Neuroscience, University of Nottingham, Clinical Sciences Building, City Hospital, Hucknall Road, Nottingham, NG5 1PB, UK
| |
Collapse
|
9
|
Tseng HW, Kulina I, Salga M, Fleming W, Vaquette C, Genêt F, Levesque JP, Alexander KA. Neurogenic Heterotopic Ossifications Develop Independently of Granulocyte Colony-Stimulating Factor and Neutrophils. J Bone Miner Res 2020; 35:2242-2251. [PMID: 32568412 DOI: 10.1002/jbmr.4118] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/05/2020] [Accepted: 06/17/2020] [Indexed: 12/25/2022]
Abstract
Neurogenic heterotopic ossifications (NHOs) are incapacitating heterotopic bones in periarticular muscles that frequently develop following traumatic brain or spinal cord injuries (SCI). Using our unique model of SCI-induced NHO, we have previously established that mononucleated phagocytes infiltrating injured muscles are required to trigger NHO via the persistent release of the pro-inflammatory cytokine oncostatin M (OSM). Because neutrophils are also a major source of OSM, we investigated whether neutrophils also play a role in NHO development after SCI. We now show that surgery transiently increased granulocyte colony-stimulating factor (G-CSF) levels in blood of operated mice, and that G-CSF receptor mRNA is expressed in the hamstrings of mice developing NHO. However, mice defective for the G-CSF receptor gene Csf3r, which are neutropenic, have unaltered NHO development after SCI compared to C57BL/6 control mice. Because the administration of recombinant human G-CSF (rhG-CSF) has been trialed after SCI to increase neuroprotection and neuronal regeneration and has been shown to suppress osteoblast function at the endosteum of skeletal bones in human and mice, we investigated the impact of a 7-day rhG-CSF treatment on NHO development. rhG-CSF treatment significantly increased neutrophils in the blood, bone marrow, and injured muscles. However, there was no change in NHO development compared to saline-treated controls. Overall, our results establish that unlike monocytes/macrophages, neutrophils are dispensable for NHO development following SCI, and rhG-CSF treatment post-SCI does not impact NHO development. Therefore, G-CSF treatment to promote neuroregeneration is unlikely to adversely promote or affect NHO development in SCI patients. © 2020 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Hsu-Wen Tseng
- Mater Research Institute, Translational Research Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | - Irina Kulina
- Mater Research Institute, Translational Research Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | - Marjorie Salga
- Mater Research Institute, Translational Research Institute, The University of Queensland, Woolloongabba, QLD, Australia.,Department of Physical Medicine and Rehabilitation, Raymond Poincaré Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Garches, France
| | - Whitney Fleming
- Mater Research Institute, Translational Research Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | - Cedryck Vaquette
- School of Dentistry, The University of Queensland, Herston, QLD, Australia.,Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD, Australia
| | - François Genêt
- Department of Physical Medicine and Rehabilitation, Raymond Poincaré Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Garches, France.,Evolution of Neuromuscular Diseases: Innovative Concepts and Practice (END:ICAP) U1179 Institut Natational de la Santé et de la Recherche Médicale, Unité de Formation et de Recherche Simone Veil-Santé, University of Versailles Saint Quentin en Yvelines, Montigny-le-Bretonneux, France
| | - Jean-Pierre Levesque
- Mater Research Institute, Translational Research Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | - Kylie A Alexander
- Mater Research Institute, Translational Research Institute, The University of Queensland, Woolloongabba, QLD, Australia
| |
Collapse
|
10
|
Sabet Sarvestani F, Azarpira N. microRNAs Alterations of Myocardium and Brain Ischemia-Reperfusion Injury: Insight to Improve Infarction. Immunol Invest 2020; 51:51-72. [DOI: 10.1080/08820139.2020.1808672] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
11
|
Tuo M, Xiao Y, Xu Y, Wang L, Wei X, Zhang L. Role of Granulocyte-colony Stimulating Factor in the Protection of Cerebral Vascular Endothelium, White Matter, and Cognition. Curr Neurovasc Res 2020; 16:425-432. [PMID: 31660819 DOI: 10.2174/1567202616666191029115113] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 09/29/2019] [Accepted: 10/02/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Granulocyte-colony stimulating factor (G-CSF) has protective effects on many neurological diseases. The effects of G-CSF on vascular endothelium and White Matter (WM) injury in Cerebral Small Vessel Disease (CSVD) were explored in this study via a model of spontaneously hypertensive rat (SHR) in order to elucidate the mechanism of G-CSF in Vascular Cognitive Impairment (VCI). METHODS 24-week-old male SHRs were randomly divided into the treatment group and model group, with the same age Wistar rats as the control group. The novel object recognition test (NORT) and Morris water maze were conducted after 7 days of G-CSF(50ug/kg) or normal saline treatment to examine their non-spatial and spatial cognitive functions. After that, a transmission electron microscope (TEM) and FLB staining were used to observe the vascular endothelial cell and WM damage. Furthermore, the expression of VEGF, MMP-9, Caspase-3, TUNEL and BrdULaminin in the cortical area was detected by immunostaining methods. RESULTS Our results showed that G-CSF promoted the expression of VEGF and BrdU+-Laminin+ endothelial cells, but down-regulated the level of MMP-9, thus significantly repaired the cerebral vascular endothelial cells and perivascular structure in SHR. The WM damage, the expression of caspase-3 and the apoptosis rate decreased after G-CSF treatment. Ultimately, G-CSF improved the non-spatial cognitive function in SHR rather than the spatial cognitive function. CONCLUSION Therefore, our findings indicated that G-CSF might facilitate the improvement of non-spatial cognitive function in CSVD by repairing endothelial cells and alleviating WM damage.
Collapse
Affiliation(s)
- Minghui Tuo
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yunyue Xiao
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yan Xu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.,Neuropsychological Research Center, Wuhan University, Hubei, 430071, China
| | - Lisha Wang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Xin Wei
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Lei Zhang
- Neuropsychological Research Center, Wuhan University, Hubei, 430071, China
| |
Collapse
|
12
|
Can miRNAs Be Considered as Diagnostic and Therapeutic Molecules in Ischemic Stroke Pathogenesis?-Current Status. Int J Mol Sci 2020; 21:ijms21186728. [PMID: 32937836 PMCID: PMC7555634 DOI: 10.3390/ijms21186728] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 09/08/2020] [Accepted: 09/10/2020] [Indexed: 12/12/2022] Open
Abstract
Ischemic stroke is one of the leading causes of death worldwide. Clinical manifestations of stroke are long-lasting and causing economic burden on the patients and society. Current therapeutic modalities to treat ischemic stroke (IS) are unsatisfactory due to the intricate pathophysiology and poor functional recovery of brain cellular compartment. MicroRNAs (miRNA) are endogenously expressed small non-coding RNA molecules, which can act as translation inhibitors and play a pivotal role in the pathophysiology associated with IS. Moreover, miRNAs may be used as potential diagnostic and therapeutic tools in clinical practice; yet, the complete role of miRNAs is enigmatic during IS. In this review, we explored the role of miRNAs in the regulation of stroke risk factors viz., arterial hypertension, metabolic disorders, and atherosclerosis. Furthermore, the role of miRNAs were reviewed during IS pathogenesis accompanied by excitotoxicity, oxidative stress, inflammation, apoptosis, angiogenesis, neurogenesis, and Alzheimer's disease. The functional role of miRNAs is a double-edged sword effect in cerebral ischemia as they could modulate pathological mechanisms associated with risk factors of IS. miRNAs pertaining to IS pathogenesis could be potential biomarkers for stroke; they could help researchers to identify a particular stroke type and enable medical professionals to evaluate the severity of brain injury. Thus, ascertaining the role of miRNAs may be useful in deciphering their diagnostic role consequently it is plausible to envisage a suitable therapeutic modality against IS.
Collapse
|
13
|
Abstract
Stroke remains a major unmet clinical need that warrants novel therapies. Following an ischemic insult, the cerebral vasculature secretes inflammatory molecules, creating the stroke vasculome profile. The present study evaluated the therapeutic effects of endothelial cells on the inflammation-associated stroke vasculome. qRT-PCR analysis revealed that specific inflammation-related vasculome genes BRM, IκB, Foxf1, and ITIH-5 significantly upregulated by oxygen glucose deprivation (OGD. Interestingly, co-culture of human endothelial cells (HEN6) with human endothelial cells (EPCs) during OGD significantly blocked the elevations of BRM, IκB, and Foxf1, but not ITIH-5. Next, employing the knockdown/antisense technology, silencing the inflammation-associated stroke vasculome gene, IκB, as opposed to scrambled knockdown, blocked the EPC-mediated protection of HEN6 against OGD. In vivo, stroke animals transplanted with intracerebral human EPCs (300,000 cells) into the striatum and cortex 4 h post ischemic stroke displayed significant behavioral recovery up to 30 days post-transplantation compared to vehicle-treated stroke animals. At 7 days post-transplantation, quantification of the fluorescent staining intensity in the cortex and striatum revealed significant upregulation of the endothelial marker RECA1 and a downregulation of the stroke-associated vasculome BRM, IKB, Foxf1, ITIH-5 and PMCA2 in the ipsilateral side of cortex and striatum of EPC-transplanted stroke animals relative to vehicle-treated stroke animals. Altogether, these results demonstrate that EPCs exert therapeutic effects in experimental stroke possibly by modulating the inflammation-plagued vasculome.
Collapse
|
14
|
Abstract
Stroke remains a major cause of serious disability due to the brain's limited capacity to regenerate. Current treatments focus on acute recanalization of the occluded blood vessels; however, currently there are no approved therapy options to regenerate neural circuits and reduce stroke-related disability. To promote recovery, therapeutic angiogenesis has been proposed as a promising target. Although restoration of blood vessels providing oxygen and nutrients to the peri-infarct regions may be beneficial, newly generated capillaries may also carry pathophysiological risk factors that need to be considered. One major concern are adverse effects including edema formation and haemorrhagic transformation due to the comprised endothelial barrier function during vascular remodelling. This brief opinion article will discuss the challenges and the newest advancements of angiogenesis as a therapeutic strategy for ischemic stroke.
Collapse
Affiliation(s)
- Ruslan Rust
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland.,Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| |
Collapse
|
15
|
Zhu L, Hu B, Guo Y, Yang H, Zheng J, Yao X, Hu H, Liu H. Effect of Chitosan oligosaccharides on ischemic symptom and gut microbiota disbalance in mice with hindlimb ischemia. Carbohydr Polym 2020; 240:116271. [PMID: 32475560 DOI: 10.1016/j.carbpol.2020.116271] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/16/2020] [Accepted: 04/07/2020] [Indexed: 02/07/2023]
Abstract
This study was designed to explore the effect of Chitosan oligosaccharides (COS) on mouse hindlimb ischemia by femoral artery ligation. Here, we demonstrated that COS treatment statistically promoted the blood perfusion and neovascularization in ischemic hindlimb of mice, accompanied by the suppression of inflammation and oxidative stress. By 16S rDNA gene sequencing, the disbalanced gut microbiota was observed in ischemic mice, while COS treatment, at least in part, restored the abundance changes of some intestinal bacteria at either phylum or genus levels. Based on metabolomics analysis on mouse plasma by UPLC-QTOF-MS, we screened 20 metabolites with the largest responses to ischemia, several of which were markedly reversed by COS. By Spearman's correlation analysis, the changed metabolites might act as a bridge between improved intestinal bacterial structure and alleviated hindlimb ischemia of mice treated by COS. Our studies point towards a potential role of COS in treatment of peripheral ischemia diseases.
Collapse
Affiliation(s)
- Lin Zhu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Baifei Hu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Yanlei Guo
- Chongqing Academy of Chinese Materia Medica, Nanshan Road 34, Chongqing 400065, PR China
| | - Huabing Yang
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Junping Zheng
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Xiaowei Yao
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Haiming Hu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China.
| | - Hongtao Liu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China; Chongqing Academy of Chinese Materia Medica, Nanshan Road 34, Chongqing 400065, PR China.
| |
Collapse
|
16
|
Kingsbury C, Heyck M, Bonsack B, Lee JY, Borlongan CV. Stroke gets in your eyes: stroke-induced retinal ischemia and the potential of stem cell therapy. Neural Regen Res 2019; 15:1014-1018. [PMID: 31823871 PMCID: PMC7034271 DOI: 10.4103/1673-5374.270293] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Stroke persists as a global health and economic crisis, yet only two interventions to reduce stroke-induced brain injury exist. In the clinic, many patients who experience an ischemic stroke often further suffer from retinal ischemia, which can inhibit their ability to make a functional recovery and may diminish their overall quality of life. Despite this, no treatments for retinal ischemia have been developed. In both cases, ischemia-induced mitochondrial dysfunction initiates a cell loss cascade and inhibits endogenous brain repair. Stem cells have the ability to transfer healthy and functional mitochondria not only ischemic neurons, but also to similarly endangered retinal cells, replacing their defective mitochondria and thereby reducing cell death. In this review, we encapsulate and assess the relationship between cerebral and retinal ischemia, recent preclinical advancements made using in vitro and in vivo retinal ischemia models, the role of mitochondrial dysfunction in retinal ischemia pathology, and the therapeutic potential of stem cell-mediated mitochondrial transfer. Furthermore, we discuss the pitfalls in classic rodent functional assessments and the potential advantages of laser Doppler as a metric of stroke progression. The studies evaluated in this review highlight stem cell-derived mitochondrial transfer as a novel therapeutic approach to both retinal ischemia and stroke. Furthermore, we posit the immense correlation between cerebral and retinal ischemia as an underserved area of study, warranting exploration with the aim of these treating injuries together.
Collapse
Affiliation(s)
- Chase Kingsbury
- Center of Excellence for Aging and Brain Repair, University of South Florida College of Medicine, Tampa, FL, USA
| | - Matt Heyck
- Center of Excellence for Aging and Brain Repair, University of South Florida College of Medicine, Tampa, FL, USA
| | - Brooke Bonsack
- Center of Excellence for Aging and Brain Repair, University of South Florida College of Medicine, Tampa, FL, USA
| | - Jea-Young Lee
- Center of Excellence for Aging and Brain Repair, University of South Florida College of Medicine, Tampa, FL, USA
| | - Cesar V Borlongan
- Center of Excellence for Aging and Brain Repair, University of South Florida College of Medicine, Tampa, FL, USA
| |
Collapse
|
17
|
Zheng ZV, Lyu H, Lam SYE, Lam PK, Poon WS, Wong GKC. The Dynamics of Microglial Polarization Reveal the Resident Neuroinflammatory Responses After Subarachnoid Hemorrhage. Transl Stroke Res 2019; 11:433-449. [PMID: 31628642 DOI: 10.1007/s12975-019-00728-5] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 08/17/2019] [Accepted: 08/20/2019] [Indexed: 01/06/2023]
Abstract
Neuroinflammation plays a critical role in the pathogenesis of subarachnoid hemorrhage (SAH). Microglia, as the resident immune cells, orchestrate neuroinflammation distinctly in neurological diseases with different polarization statuses. However, microglial polarizations in the neuroinflammatory responses after SAH are not fully understood. In this study, we investigated the dynamics of microglial reaction in an endovascular perforated SAH model. By using the Cx3cr1GFP/GFP Ccr2RFP/RFP transgenic mice, we found that the reactive immune cells were largely from resident microglia pool rather than infiltrating macrophages. Immunostaining and real-time PCR were employed to analyze the temporal microglial polarization and the resulting inflammatory responses. Our results showed that microglia accumulated immediately after SAH with a centrifugal spreading through the Cortex Adjacent to the Perforated Site (CAPS) to the remote motor cortex. Microglia polarized dynamically from M1 to M2 phenotype along with the morphological transformation from ramified to amoeboid shapes. The ramified microglia demonstrated the M1 property, which suggested the function-related microglial polarization occurred prior to morphological transformation after SAH. Bipolar-shaped microglia appeared as the intermediate and transitional status with the capacity of bidirectional polarization. The microglial polarization status is distinct in molecular inflammatory responses; M1-related pro-inflammation was predominant in the early phase and subsequently transited to the M2-related anti-inflammation. The systematic characterization of the dynamics of microglial polarization in this study contributes to the understanding of the origin of neuroinflammatory responses after SAH and provides key foundation for further investigations to develop target treatment.
Collapse
Affiliation(s)
- Zhiyuan Vera Zheng
- Division of Neurosurgery, Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Hao Lyu
- Division of Neurosurgery, Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Sin Yu Erica Lam
- Division of Neurosurgery, Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Ping Kuen Lam
- Division of Neurosurgery, Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Wai Sang Poon
- Division of Neurosurgery, Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - George Kwok Chu Wong
- Division of Neurosurgery, Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China. .,Department of Surgery, Prince of Wales Hospital, 4/F, Lui Che Woo Clinical Sciences Building, 30-32 Ngan Shing Street, Shatin, NT, Hong Kong, China.
| |
Collapse
|
18
|
Zhang H, Lee JY, Borlongan CV, Tajiri N. A brief physical activity protects against ischemic stroke. Brain Circ 2019; 5:112-118. [PMID: 31620657 PMCID: PMC6785942 DOI: 10.4103/bc.bc_32_19] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 08/31/2019] [Accepted: 09/07/2019] [Indexed: 02/07/2023] Open
Abstract
With restricted therapeutic opportunities, stroke remains a relevant, critical disease necessitating study. Due to the unique aspect of ischemic strokes, finding approaches to maintain the vigor of the cerebral vasculature, such as increased angiogenesis, may protect against stroke. Ischemic strokes are caused by disruptions in blood movement in the brain, resulting in a torrent of harmful cerebrovasculature modifications. In an investigation by Pianta et al., Sprague-Dawley rats have been separated into those that undergo exercise prior to middle cerebral artery occlusion (MCAO) and those that were not exposed to physical activity preceding MCAO. The outcomes and results of the current study gave new insights into the capacity of exercise to help prevent ischemic strokes or mitigate poststroke effects. The data collected from the study suggested that rats that went through a short bout of exercise before MCAO presented superior motor performance, more active cells in the peri-infarct region, and reduced infarct sizes. When compared to the control group, the rats that went through exercise also had heightened angiogenesis and improved neuroprotection. Thus, a brief bout of physical activity preceding a stroke may provide neuroprotection by enhancing the strength of the cerebrovasculature in the brain. This notion that even an instant of physical exercise before a stroke is induced can help dampen the effects of ischemic stroke, which could lead to future techniques in preventing the ischemic stroke so that it never happens at all.
Collapse
Affiliation(s)
- Henry Zhang
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Jea-Young Lee
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, 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, Tampa, FL, USA
| | - Naoki Tajiri
- Department of Neurophysiology and Brain Science, Graduate School of Medical Sciences and Medical School, Nagoya City University, Nagoya, Japan
| |
Collapse
|
19
|
Tuazon JP, Castelli V, Borlongan CV. Drug-like delivery methods of stem cells as biologics for stroke. Expert Opin Drug Deliv 2019; 16:823-833. [PMID: 31311344 DOI: 10.1080/17425247.2019.1645116] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Introduction: Stem cell therapy is an experimental treatment for brain disorders. Although a cellular product, stem cells can be classified as biologics based on the cells' secretion of therapeutic substances. Treatment with stem cell biologics may appeal to stroke because of the secondary cell death mechanisms, especially neuroinflammation, that are rampant from the onset and remain elevated during the progressive phase of the disease requiring multi-pronged biological targets to effectively abrogate the neurodegenerative pathology. However, the optimal delivery methods, among other logistical approaches (i.e. cell doses and timing of intervention), for stem cell therapy will need to be refined before stem cell biologics can be successfully utilized for stroke in large scale clinical trials. Areas covered: In this review, we discuss how the innate qualities of stem cells characterize them as biologics, how stem cell transplantation may be an ideal treatment for stroke, and the various routes of stem cell administration that have been employed in various preclinical and clinical investigations. Expert opinion: There is a need to optimize the delivery of stem cell biologics for stroke in order to guide the safe and effective translation of this therapy from the laboratory to the clinic.
Collapse
Affiliation(s)
- Julian P Tuazon
- a Center of Excellence for Aging & Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine , Tampa , FL , USA
| | - Vanessa Castelli
- a Center of Excellence for Aging & Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine , Tampa , FL , USA
| | - Cesar V Borlongan
- a Center of Excellence for Aging & Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine , Tampa , FL , USA
| |
Collapse
|
20
|
Kim JS. tPA Helpers in the Treatment of Acute Ischemic Stroke: Are They Ready for Clinical Use? J Stroke 2019; 21:160-174. [PMID: 31161761 PMCID: PMC6549064 DOI: 10.5853/jos.2019.00584] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 04/15/2019] [Indexed: 12/12/2022] Open
Abstract
Tissue plasminogen activator (tPA) is the only therapeutic agent approved to treat patients with acute ischemic stroke. The clinical benefits of tPA manifest when the agent is administered within 4.5 hours of stroke onset. However, tPA administration, especially delayed administration, is associated with increased intracranial hemorrhage (ICH), hemorrhagic transformation (HT), and mortality. In the ischemic brain, vascular remodeling factors are upregulated and microvascular structures are destabilized. These factors disrupt the blood brain barrier (BBB). Delayed recanalization of the vessels in the presence of relatively matured infarction appears to damage the BBB, resulting in HT or ICH, also known as reperfusion injury. Moreover, tPA itself activates matrix metalloproteases, further aggravating BBB disruption. Therefore, attenuation of edema, HT, or ICH after tPA treatment is an important therapeutic strategy that may enable clinicians to extend therapeutic time and increase the probability of excellent outcomes. Recently, numerous agents with various mechanisms have been developed to interfere with various steps of ischemia/ reperfusion injuries or BBB destabilization. These agents successfully reduce infarct volume and decrease the incidence of ICH and HT after delayed tPA treatment in various animal stroke models. However, only some have entered into clinical trials; the results have been intriguing yet unsatisfactory. In this narrative review, I describe such drugs and discuss the problems and future directions. These “tPA helpers” may be clinically used in the future to increase the efficacy of tPA in patients with acute ischemic stroke.
Collapse
Affiliation(s)
- Jong S Kim
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
21
|
Ouk T, Potey C, Maestrini I, Petrault M, Mendyk AM, Leys D, Bordet R, Gautier S. Neutrophils in tPA-induced hemorrhagic transformations: Main culprit, accomplice or innocent bystander? Pharmacol Ther 2019; 194:73-83. [DOI: 10.1016/j.pharmthera.2018.09.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
22
|
Dynamic Detection of Thrombolysis in Embolic Stroke Rats by Synchrotron Radiation Angiography. Transl Stroke Res 2019; 10:695-704. [PMID: 30680639 DOI: 10.1007/s12975-019-0687-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 11/30/2018] [Accepted: 01/03/2019] [Indexed: 12/20/2022]
Abstract
A rodent model of embolic middle cerebral artery occlusion is used to mimic cerebral embolism in clinical patients. Thrombolytic therapy is the effective treatment for this ischemic injury. However, it is difficult to detect thrombolysis dynamically in living animals. Synchrotron radiation angiography may provide a novel approach to directly monitor the thrombolytic process and assess collateral circulation after embolic stroke. Thirty-six adult Sprague-Dawley rats underwent the embolic stroke model procedure and were then treated with tissue plasminogen activator. The angiographic images were obtained in vivo by synchrotron radiation angiography. Synchrotron radiation angiography confirmed the successful establishment of occlusion and detected the thrombolysis process after the thrombolytic treatment. The time of thrombolytic recanalization was unstable during embolic stroke. The infarct volume increased as the recanalization time was delayed from 2 to 6 h (p < 0.05). The collateral circulation of the internal carotid artery to the ophthalmic artery, the olfactory artery to the ophthalmic artery, and the posterior cerebral artery to the middle cerebral artery opened after embolic stroke and manifested different opening rates (59%, 24%, and 75%, respectively) in the rats. The opening of the collateral circulation from the posterior cerebral artery to the middle cerebral artery alleviated infarction in rats with successful thrombolysis (p < 0.05). The cerebral vessels of the circle of Willis narrowed after thrombolysis (p < 0.05). Synchrotron radiation angiography provided a unique tool to dynamically detect and assess the thrombolysis process and the collateral circulation during thrombolytic therapy.
Collapse
|
23
|
Incontri Abraham D, Gonzales M, Ibarra A, Borlongan CV. Stand alone or join forces? Stem cell therapy for stroke. Expert Opin Biol Ther 2018; 19:25-33. [PMID: 30477353 DOI: 10.1080/14712598.2019.1551872] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Stroke is a major cause of mortality and disability with a narrow therapeutic window. Stem cell therapy may enhance the stroke recovery. AREAS COVERE Regenerative medicine via stem cells stands as a novel therapy for stroke. In particular, bone marrow-derived mesenchymal stem cells (MSCs) have neuroprotective and anti-inflammatory properties that improve brain function after stroke. Here, we discuss the safety, efficacy, and mechanism of action underlying the therapeutic effects of bone marrow-derived MSCs. We also examine the discrepant transplant protocols between preclinical studies and clinical trials. Laboratory studies show the safety and efficacy of bone marrow-derived MSCs in stroke models. However, while safe, MSCs remain to be fully evaluated as effective in clinical trials. Furthermore, recognizing the multiple cell death processes associated with stroke, we next discuss the potential therapeutic benefits of a combination therapy. With preliminary results and on-going clinical trials, a careful assessment of dosing, timing, and delivery route regimens will further direct the future of stem cell therapy for neurological disorders, including stroke. EXPERT OPINION Bone marrow-derived MSCs appear to be the optimal stem cell source for stroke therapy. Optimizing dosing, timing, and delivery route should guide the clinical application of bone marrow-derived MSCs.
Collapse
Affiliation(s)
- Diego Incontri Abraham
- a Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair , University of South Florida Morsani College of Medicine , Tampa , FL , USA.,b Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud , Universidad Anáhuac México Campus Norte , Huixquilucan, Edo. de Mexico , México
| | - Melissa Gonzales
- a Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair , University of South Florida Morsani College of Medicine , Tampa , FL , USA
| | - Antonio Ibarra
- b Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud , Universidad Anáhuac México Campus Norte , Huixquilucan, Edo. de Mexico , México.,c Faculty of Health Sciences , Proyecto CAMINA A.C , Ciudad de México , México
| | - Cesar V Borlongan
- a Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair , University of South Florida Morsani College of Medicine , Tampa , FL , USA
| |
Collapse
|
24
|
Zhang Y, Ma L, Ren C, Liu K, Tian X, Wu D, Ding Y, Li J, Borlongan CV, Ji X. Immediate remote ischemic postconditioning reduces cerebral damage in ischemic stroke mice by enhancing leptomeningeal collateral circulation. J Cell Physiol 2018; 234:12637-12645. [PMID: 30536714 PMCID: PMC6590306 DOI: 10.1002/jcp.27858] [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: 10/02/2018] [Accepted: 11/15/2018] [Indexed: 12/22/2022]
Abstract
Remote ischemic postconditioning (RIPC) is a promising neuroprotective strategy for ischemic stroke. Here, we employed a focal ischemic stroke mouse model to test the hypothesis that poststroke collateral circulation as a potent mechanism of action underlying the therapeutic effects of immediate RIPC. During reperfusion of cerebral ischemia, the mice were randomly assigned to receive RIPC, granulocyte colony‐stimulating factor (G‐CSF) as a positive control, or no treatment. At 24 hr, we found RIPC and G‐CSF increased monocytes/macrophages in the dorsal brain surface and in the spleen, coupled with enhanced leptomeningeal collateral flow compared with nontreatment group. Blood monocytes depletion by 5‐fluorouracil (5‐FU) significantly limited the neuroprotection of RIPC or G‐CSF treatment. The protein expression of proangiogenic factors such as Ang‐2 was increased by ischemia, but treatment with either RIPC or G‐CSF showed no further upregulation. Thus, immediate RIPC confers neuroprotection, in part, by enhancing leptomeningeal collateral circulation in a mouse model of ischemic stroke.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Neurobiology, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Longhui Ma
- Department of Neurobiology, Capital Medical University, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Changhong Ren
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Kaiyin Liu
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan
| | - Xin Tian
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Di Wu
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan
| | - Junfa Li
- Department of Neurobiology, Capital Medical University, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Cesar V Borlongan
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, Florida
| | - Xunming Ji
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China.,Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
25
|
Thiebaut AM, Gauberti M, Ali C, Martinez De Lizarrondo S, Vivien D, Yepes M, Roussel BD. The role of plasminogen activators in stroke treatment: fibrinolysis and beyond. Lancet Neurol 2018; 17:1121-1132. [PMID: 30507392 DOI: 10.1016/s1474-4422(18)30323-5] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 07/25/2018] [Accepted: 08/28/2018] [Indexed: 12/20/2022]
Abstract
Although recent technical advances in thrombectomy have revolutionised acute stroke treatment, prevalence of disability and death related to stroke remain high. Therefore, plasminogen activators-eukaryotic, bacterial, or engineered forms that can promote fibrinolysis by converting plasminogen into active plasmin and facilitate clot breakdown-are still commonly used in the acute treatment of ischaemic stroke. Hence, plasminogen activators have become a crucial area for clinical investigation for their ability to recanalise occluded arteries in ischaemic stroke and to accelerate haematoma clearance in haemorrhagic stroke. However, inconsistent results, insufficient evidence of efficacy, or reports of side-effects in trial settings might reduce the use of plasminogen activators in clinical practice. Additionally, the mechanism of action for plasminogen activators could extend beyond the vessel lumen and involve plasminogen-independent processes, which would suggest that plasminogen activators have also non-fibrinolytic roles. Understanding the complex mechanisms of action of plasminogen activators can guide future directions for therapeutic interventions in patients with stroke.
Collapse
Affiliation(s)
- Audrey M Thiebaut
- Normandie Université, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Cyceron, Caen, France
| | - Maxime Gauberti
- Normandie Université, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Cyceron, Caen, France
| | - Carine Ali
- Normandie Université, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Cyceron, Caen, France
| | - Sara Martinez De Lizarrondo
- Normandie Université, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Cyceron, Caen, France
| | - Denis Vivien
- Normandie Université, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Cyceron, Caen, France; Clinical Research Department, University Hospital Caen-Normandy, Caen, France
| | - Manuel Yepes
- Department of Neurology and Center for Neurodegenerative Disease, Emory University School of Medicine, Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, and Department of Neurology, Veterans Affairs Medical Center, Atlanta, GA, USA
| | - Benoit D Roussel
- Normandie Université, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Cyceron, Caen, France.
| |
Collapse
|
26
|
Meadows KL. Experimental models of focal and multifocal cerebral ischemia: a review. Rev Neurosci 2018; 29:661-674. [PMID: 29397392 DOI: 10.1515/revneuro-2017-0076] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 11/26/2017] [Indexed: 02/07/2023]
Abstract
Rodent and rabbit stroke models have been instrumental in our current understanding of stroke pathophysiology; however, translational failure is a significant problem in preclinical ischemic stroke research today. There are a number of different focal cerebral ischemia models that vary in their utility, pathophysiology of causing disease, and their response to treatments. Unfortunately, despite active preclinical research using these models, treatment options for ischemic stroke have not significantly advanced since the food and drug administration approval of tissue plasminogen activator in 1996. This review aims to summarize current stroke therapies, the preclinical experimental models used to help develop stroke therapies, as well as their advantages and limitations. In addition, this review discusses the potential for naturally occurring canine ischemic stroke models to compliment current preclinical models and to help bridge the translational gap between small mammal models and human clinical trials.
Collapse
Affiliation(s)
- Kristy L Meadows
- Cummings School of Veterinary Medicine, Tufts University, 200 Westboro Road, Grafton, MA 01536, USA
| |
Collapse
|
27
|
Knecht T, Borlongan C, Dela Peña I. Combination therapy for ischemic stroke: Novel approaches to lengthen therapeutic window of tissue plasminogen activator. Brain Circ 2018; 4:99-108. [PMID: 30450415 PMCID: PMC6187940 DOI: 10.4103/bc.bc_21_18] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 08/29/2018] [Accepted: 09/10/2018] [Indexed: 12/24/2022] Open
Abstract
Tissue plasminogen activator (tPA) thrombolysis continues to be the gold standard therapy for ischemic stroke. Due to the time-limited treatment window, within 4.5 h of stroke onset, and a variety of potentially deadly complications related to delayed administration, particularly hemorrhagic transformation (HT), clinical use of tPA is limited. Combination therapies with other interventions, drug or nondrug, have been hypothesized as a logical approach to enhancing tPA effectiveness. Here, we discuss various potential pharmacological and nondrug treatments to minimize adverse effects, primarily HT, associated with delayed tPA administration. Pharmacological interventions include many that support the integrity of the blood–brain barrier (i.e., atorvastatin, batimastat, candesartan, cilostazol, fasudil, and minocycline), promote vascularization and preserve cerebrovasculature (i.e., coumarin derivative IMM-H004 and granulocyte-colony stimulating factor), employing other mechanisms of action (i.e., oxygen transporters and ascorbic acid). Nondrug treatments are comprised of stem cell transplantation and gas therapies with multi-faceted approaches. Combination therapy with tPA and the aforementioned treatments demonstrated promise for mitigating the adverse complications associated with delayed tPA treatment and rescuing stroke-induced behavioral deficits. Therefore, the conjunctive therapy method is a novel therapeutic approach that can attempt to minimize the limitations of tPA treatment and possibly increase the therapeutic window for ischemic stroke treatment.
Collapse
Affiliation(s)
- Talia Knecht
- Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Loma Linda University, Loma Linda, CA, USA
| | - Cesar Borlongan
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida College of Medicine, Tampa, Florida, USA
| | - Ike Dela Peña
- Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
28
|
Shekhar S, Cunningham MW, Pabbidi MR, Wang S, Booz GW, Fan F. Targeting vascular inflammation in ischemic stroke: Recent developments on novel immunomodulatory approaches. Eur J Pharmacol 2018; 833:531-544. [PMID: 29935175 DOI: 10.1016/j.ejphar.2018.06.028] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/02/2018] [Accepted: 06/19/2018] [Indexed: 10/28/2022]
Abstract
Ischemic stroke is a devastating and debilitating medical condition with limited therapeutic options. However, accumulating evidence indicates a central role of inflammation in all aspects of stroke including its initiation, the progression of injury, and recovery or wound healing. A central target of inflammation is disruption of the blood brain barrier or neurovascular unit. Here we discuss recent developments in identifying potential molecular targets and immunomodulatory approaches to preserve or protect barrier function and limit infarct damage and functional impairment. These include blocking harmful inflammatory signaling in endothelial cells, microglia/macrophages, or Th17/γδ T cells with biologics, third generation epoxyeicosatrienoic acid (EET) analogs with extended half-life, and miRNA antagomirs. Complementary beneficial pathways may be enhanced by miRNA mimetics or hyperbaric oxygenation. These immunomodulatory approaches could be used to greatly expand the therapeutic window for thrombolytic treatment with tissue plasminogen activator (t-PA). Moreover, nanoparticle technology allows for the selective targeting of endothelial cells for delivery of DNA/RNA oligonucleotides and neuroprotective drugs. In addition, although likely detrimental to the progression of ischemic stroke by inducing inflammation, oxidative stress, and neuronal cell death, 20-HETE may also reduce susceptibility of onset of ischemic stroke by maintaining autoregulation of cerebral blood flow. Although the interaction between inflammation and stroke is multifaceted, a better understanding of the mechanisms behind the pro-inflammatory state at all stages will hopefully help in developing novel immunomodulatory approaches to improve mortality and functional outcome of those inflicted with ischemic stroke.
Collapse
Affiliation(s)
- Shashank Shekhar
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS, USA; Institute of Clinical Medicine, University of Turku, Turku, Finland
| | - Mark W Cunningham
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Mallikarjuna R Pabbidi
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Shaoxun Wang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - George W Booz
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Fan Fan
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA.
| |
Collapse
|
29
|
Dela Peña IC, Yang S, Shen G, Fang Liang H, Solak S, Borlongan CV. Extension of Tissue Plasminogen Activator Treatment Window by Granulocyte-Colony Stimulating Factor in a Thromboembolic Rat Model of Stroke. Int J Mol Sci 2018; 19:ijms19061635. [PMID: 29857523 PMCID: PMC6032420 DOI: 10.3390/ijms19061635] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 05/27/2018] [Accepted: 05/29/2018] [Indexed: 12/26/2022] Open
Abstract
When given beyond 4.5 h of stroke onset, tissue plasminogen activator (tPA) induces deleterious side effects in the ischemic brain, notably, hemorrhagic transformation (HT). We examined the efficacy of granulocyte-colony stimulating factor (G-CSF) in reducing delayed tPA-induced HT, cerebral infarction, and neurological deficits in a thromboembolic (TE) stroke model, and whether the effects of G-CSF were sustained for longer periods of recovery. After stroke induction, rats were given intravenous saline (control), tPA (10 mg/kg), or G-CSF (300 μg/kg) + tPA 6 h after stroke. We found that G-CSF reduced delayed tPA-associated HT by 47%, decreased infarct volumes by 33%, and improved motor and neurological deficits by 15% and 25%, respectively. It also prevented delayed tPA treatment-induced mortality by 46%. Immunohistochemistry showed 1.5- and 1.8-fold enrichment of the endothelial progenitor cell (EPC) markers CD34+ and VEGFR2 in the ischemic cortex and striatum, respectively, and 1.7- and 2.8-fold increases in the expression of the vasculogenesis marker von Willebrand factor (vWF) in the ischemic cortex and striatum, respectively, in G-CSF-treated rats compared with tPA-treated animals. Flow cytometry revealed increased mobilization of CD34+ cells in the peripheral blood of rats given G-CSF. These results corroborate the efficacy of G-CSF in enhancing the therapeutic time window of tPA for stroke treatment via EPC mobilization and enhancement of vasculogenesis.
Collapse
Affiliation(s)
- Ike C Dela Peña
- Department of Pharmaceutical and Administrative Sciences, Loma Linda University, Loma Linda, CA 92350, USA.
| | - Samuel Yang
- Department of Pharmaceutical and Administrative Sciences, Loma Linda University, Loma Linda, CA 92350, USA.
| | - Guofang Shen
- Department of Pharmaceutical and Administrative Sciences, Loma Linda University, Loma Linda, CA 92350, USA.
| | - Hsiao Fang Liang
- Department of Pharmaceutical and Administrative Sciences, Loma Linda University, Loma Linda, CA 92350, USA.
| | - Sara Solak
- Department of Pharmaceutical and Administrative Sciences, Loma Linda University, Loma Linda, CA 92350, 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, Tampa, FL 33612, USA.
| |
Collapse
|
30
|
Wei M, Lyu H, Huo K, Su H. Impact of Bone Fracture on Ischemic Stroke Recovery. Int J Mol Sci 2018; 19:ijms19051533. [PMID: 29786644 PMCID: PMC5983742 DOI: 10.3390/ijms19051533] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 05/17/2018] [Accepted: 05/18/2018] [Indexed: 01/21/2023] Open
Abstract
Stroke is one of the most devastating complications of bone fracture, occurring in up to 4% of patients after surgical repair for hip fracture. Bone fracture and ischemic stroke have many common risk factors. The impact of bone fracture on stroke recovery has not drawn much attention in the research field. Bone fracture could occur in stroke patients at different times during the recovery phase, which steepens the trajectory of cognitive decline, greatly affects the quality of life, and causes a heavy burden on healthcare resources. In this paper, we reviewed the growing information on the pathophysiological mechanisms by which bone fracture may affect ischemic stroke recovery process.
Collapse
Affiliation(s)
- Meng Wei
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94110, USA.
| | - Haiyian Lyu
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94110, USA.
| | - Kang Huo
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94110, USA.
| | - Hua Su
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94110, USA.
| |
Collapse
|
31
|
Mizuma A, You JS, Yenari MA. Targeting Reperfusion Injury in the Age of Mechanical Thrombectomy. Stroke 2018; 49:1796-1802. [PMID: 29760275 DOI: 10.1161/strokeaha.117.017286] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Atsushi Mizuma
- From the Department of Neurology, University of California, San Francisco (A.M., J.S.Y., M.A.Y.).,San Francisco Veterans Affairs Medical Center, CA (A.M., J.S.Y., M.A.Y.).,Department of Neurology, Tokai University School of Medicine, Isehara, Japan (A.M.)
| | - Je Sung You
- From the Department of Neurology, University of California, San Francisco (A.M., J.S.Y., M.A.Y.).,San Francisco Veterans Affairs Medical Center, CA (A.M., J.S.Y., M.A.Y.).,Department of Emergency Medicine, Yonsei University College of Medicine, Seoul, South Korea (J.S.Y.)
| | - Midori A Yenari
- From the Department of Neurology, University of California, San Francisco (A.M., J.S.Y., M.A.Y.) .,San Francisco Veterans Affairs Medical Center, CA (A.M., J.S.Y., M.A.Y.)
| |
Collapse
|
32
|
Li G, Morris-Blanco KC, Lopez MS, Yang T, Zhao H, Vemuganti R, Luo Y. Impact of microRNAs on ischemic stroke: From pre- to post-disease. Prog Neurobiol 2018; 163-164:59-78. [DOI: 10.1016/j.pneurobio.2017.08.002] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 06/12/2017] [Accepted: 08/16/2017] [Indexed: 12/21/2022]
|
33
|
Ren W, Yang X. Pathophysiology of Long Non-coding RNAs in Ischemic Stroke. Front Mol Neurosci 2018; 11:96. [PMID: 29651234 PMCID: PMC5884949 DOI: 10.3389/fnmol.2018.00096] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 03/12/2018] [Indexed: 12/12/2022] Open
Abstract
Stroke is a neurological disease with high disability and fatality rates, and ischemic stroke accounts for 75% of all stroke cases. The underlying pathophysiologic processes of ischemic stroke include oxidative stress, toxicity of excitatory amino acids, excess calcium ions, increased apoptosis and inflammation. Long non-coding RNAs (lncRNAs) may participate in the regulation of the pathophysiologic processes of ischemic stroke as indicated by altered expression of lncRNAs in blood samples of acute ischemic stroke patients, animal models of focal cerebral ischemia and oxygen-glucose deprivation (OGD) cell models. Because of the potentially important role, lncRNAs might be useful as biomarkers for the diagnosis, treatment and prognosis of ischemic stroke. This article reviews the functions of lncRNAs in different pathophysiology events of ischemic stroke with a focus on specific lncRNAs that may underlie ischemic stroke pathophysiology and that could therefore serve as potential diagnostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Weimin Ren
- Center Laboratory, Jinshan Hospital, Fudan University, Shanghai, China
| | - Xiaobo Yang
- Department of Neurology, Jinshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
34
|
Dong MX, Li CM, Shen P, Hu QC, Wei YD, Ren YF, Yu J, Gui SW, Liu YY, Pan JX, Xie P. Recombinant tissue plasminogen activator induces long-term anxiety-like behaviors via the ERK1/2-GAD1-GABA cascade in the hippocampus of a rat model. Neuropharmacology 2018; 128:119-131. [DOI: 10.1016/j.neuropharm.2017.09.039] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 09/26/2017] [Accepted: 09/30/2017] [Indexed: 01/04/2023]
|
35
|
Adjunctive Therapy Approaches for Ischemic Stroke: Innovations to Expand Time Window of Treatment. Int J Mol Sci 2017; 18:ijms18122756. [PMID: 29257093 PMCID: PMC5751355 DOI: 10.3390/ijms18122756] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 12/12/2017] [Accepted: 12/16/2017] [Indexed: 12/18/2022] Open
Abstract
Tissue plasminogen activator (tPA) thrombolysis remains the gold standard treatment for ischemic stroke. A time-constrained therapeutic window, with the drug to be given within 4.5 h after stroke onset, and lethal side effects associated with delayed treatment, most notably hemorrhagic transformation (HT), limit the clinical use of tPA. Co-administering tPA with other agents, including drug or non-drug interventions, has been proposed as a practical strategy to address the limitations of tPA. Here, we discuss the pharmacological and non-drug approaches that were examined to mitigate the complications-especially HT-associated with delayed tPA treatment. The pharmacological treatments include those that preserve the blood-brain barrier (e.g., atovarstatin, batimastat, candesartan, cilostazol, fasudil, minocycline, etc.), enhance vascularization and protect the cerebrovasculature (e.g., coumarin derivate IMM-H004 and granulocyte-colony stimulating factor (G-CSF)), and exert their effects through other modes of action (e.g., oxygen transporters, ascorbic acid, etc.). The non-drug approaches include stem cell treatments and gas therapy with multi-pronged biological effects. Co-administering tPA with the abovementioned therapies showed promise in attenuating delayed tPA-induced side effects and stroke-induced neurological and behavioral deficits. Thus, adjunctive treatment approach is an innovative therapeutic modality that can address the limitations of tPA treatment and potentially expand the time window for ischemic stroke therapy.
Collapse
|
36
|
Oxidative stress and DNA damage after cerebral ischemia: Potential therapeutic targets to repair the genome and improve stroke recovery. Neuropharmacology 2017; 134:208-217. [PMID: 29128308 DOI: 10.1016/j.neuropharm.2017.11.011] [Citation(s) in RCA: 184] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 11/02/2017] [Accepted: 11/05/2017] [Indexed: 12/12/2022]
Abstract
The past two decades have witnessed remarkable advances in oxidative stress research, particularly in the context of ischemic brain injury. Oxidative stress in ischemic tissues compromises the integrity of the genome, resulting in DNA lesions, cell death in neurons, glial cells, and vascular cells, and impairments in neurological recovery after stroke. As DNA is particularly vulnerable to oxidative attack, cells have evolved the ability to induce multiple DNA repair mechanisms, including base excision repair (BER), nucleotide excision repair (NER) and non-homogenous endpoint jointing (NHEJ). Defective DNA repair is tightly correlated with worse neurological outcomes after stroke, whereas upregulation of DNA repair enzymes, such as APE1, OGG1, and XRCC1, improves long-term functional recovery following stroke. Indeed, DNA damage and repair are now known to play critical roles in fundamental aspects of stroke recovery, such as neurogenesis, white matter recovery, and neurovascular unit remodeling. Several DNA repair enzymes are essential for comprehensive neural repair mechanisms after stroke, including Polβ and NEIL3 for neurogenesis, APE1 for white matter repair, Gadd45b for axonal regeneration, and DNA-PKs for neurovascular remodeling. This review discusses the emerging role of DNA damage and repair in functional recovery after stroke and highlights the contribution of DNA repair to regenerative elements after stroke. This article is part of the Special Issue entitled 'Cerebral Ischemia'.
Collapse
|
37
|
Cai M, Zhang W, Weng Z, Stetler RA, Jiang X, Shi Y, Gao Y, Chen J. Promoting Neurovascular Recovery in Aged Mice after Ischemic Stroke - Prophylactic Effect of Omega-3 Polyunsaturated Fatty Acids. Aging Dis 2017; 8:531-545. [PMID: 28966799 PMCID: PMC5614319 DOI: 10.14336/ad.2017.0520] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 05/20/2017] [Indexed: 12/17/2022] Open
Abstract
The aged population is among the highest at risk for ischemic stroke, yet most stroke patients of advanced ages (>80 years) are excluded from access to thrombolytic treatment by tissue plasminogen activator, the only FDA approved pharmacological therapy for stroke victims. Omega-3 polyunsaturated fatty acids (n-3 PUFAs) robustly alleviate ischemic brain injury in young adult rodents, but have not yet been studied in aged animals. This study investigated whether chronic dietary supplementation of n-3 PUFAs protects aging brain against cerebral ischemia and improves long-term neurological outcomes. Aged (18-month-old) mice were administered n-3 PUFA-enriched fish oil in daily chow for 3 months before and up to 8 weeks after 45 minutes of transient middle cerebral artery occlusion (tMCAO). Sensorimotor outcomes were assessed by cylinder test and corner test up to 35 days and brain repair dynamics evaluated immunohistologically up to 56 days after tMCAO. Mice receiving dietary supplementation of n-3 PUFAs for 3 months showed significant increases in brain ratio of n-3/n-6 PUFA contents, and markedly reduced long-term sensorimotor deficits and chronic ischemic brain tissue loss after tMCAO. Mechanistically, n-3 PUFAs robustly promoted post-ischemic angiogenesis and neurogenesis, and enhanced white matter integrity after tMCAO. The Pearson linear regression analysis revealed that the enhancement of neurogenesis and white matter integrity both correlated positively with improved sensorimotor activities after tMCAO. This study demonstrates that prophylactic dietary supplementation of n-3 PUFAs effectively improves long-term stroke outcomes in aged mice, perhaps by promoting post-stroke brain repair processes such as angiogenesis, neurogenesis, and white matter restoration.
Collapse
Affiliation(s)
- Mengfei Cai
- 1State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, and Collaborative Innovation Center, Fudan University, Shanghai 200032, China
| | - Wenting Zhang
- 1State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, and Collaborative Innovation Center, Fudan University, Shanghai 200032, China
| | - Zhongfang Weng
- 2Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - R Anne Stetler
- 1State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, and Collaborative Innovation Center, Fudan University, Shanghai 200032, China.,2Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.,3Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA
| | - Xiaoyan Jiang
- 2Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Yejie Shi
- 2Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.,3Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA
| | - Yanqin Gao
- 1State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, and Collaborative Innovation Center, Fudan University, Shanghai 200032, China.,2Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Jun Chen
- 1State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, and Collaborative Innovation Center, Fudan University, Shanghai 200032, China.,2Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.,3Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA
| |
Collapse
|
38
|
Mizuma A, Yenari MA. Anti-Inflammatory Targets for the Treatment of Reperfusion Injury in Stroke. Front Neurol 2017; 8:467. [PMID: 28936196 PMCID: PMC5594066 DOI: 10.3389/fneur.2017.00467] [Citation(s) in RCA: 162] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 08/23/2017] [Indexed: 12/20/2022] Open
Abstract
While the mainstay of acute stroke treatment includes revascularization via recombinant tissue plasminogen activator or mechanical thrombectomy, only a minority of stroke patients are eligible for treatment, as delayed treatment can lead to worsened outcome. This worsened outcome at the experimental level has been attributed to an entity known as reperfusion injury (R/I). R/I is occurred when revascularization is delayed after critical brain and vascular injury has occurred, so that when oxygenated blood is restored, ischemic damage is increased, rather than decreased. R/I can increase lesion size and also worsen blood barrier breakdown and lead to brain edema and hemorrhage. A major mechanism underlying R/I is that of poststroke inflammation. The poststroke immune response consists of the aberrant activation of glial cell, infiltration of peripheral leukocytes, and the release of damage-associated molecular pattern (DAMP) molecules elaborated by ischemic cells of the brain. Inflammatory mediators involved in this response include cytokines, chemokines, adhesion molecules, and several immune molecule effectors such as matrix metalloproteinases-9, inducible nitric oxide synthase, nitric oxide, and reactive oxygen species. Several experimental studies over the years have characterized these molecules and have shown that their inhibition improves neurological outcome. Yet, numerous clinical studies failed to demonstrate any positive outcomes in stroke patients. However, many of these clinical trials were carried out before the routine use of revascularization therapies. In this review, we cover mechanisms of inflammation involved in R/I, therapeutic targets, and relevant experimental and clinical studies, which might stimulate renewed interest in designing clinical trials to specifically target R/I. We propose that by targeting anti-inflammatory targets in R/I as a combined therapy, it may be possible to further improve outcomes from pharmacological thrombolysis or mechanical thrombectomy.
Collapse
Affiliation(s)
- Atsushi Mizuma
- Department of Neurology, University of California, San Francisco and Veterans Affairs Medical Center, San Francisco, CA, United States
| | - Midori A Yenari
- Department of Neurology, University of California, San Francisco and Veterans Affairs Medical Center, San Francisco, CA, United States
| |
Collapse
|
39
|
CXCL12 enhances angiogenesis through CXCR7 activation in human umbilical vein endothelial cells. Sci Rep 2017; 7:8289. [PMID: 28811579 PMCID: PMC5557870 DOI: 10.1038/s41598-017-08840-y] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 07/13/2017] [Indexed: 12/20/2022] Open
Abstract
Angiogenesis is the process by which new vessels form from existing vascular networks. Human umbilical vein endothelial cells (HUVECs) may contribute to the study of vascular repair and angiogenesis. The chemokine CXCL12 regulates multiple cell functions, including angiogenesis, mainly through its receptor CXCR4. In contrast to CXCL12/CXCR4, few studies have described roles for CXCR7 in vascular biology, and the downstream mechanism of CXCR7 in angiogenesis remains unclear. The results of the present study showed that CXCL12 dose-dependently enhanced angiogenesis in chorioallantoic membranes (CAMs) and HUVECs. The specific activation of CXCR7 with TC14012 (a CXCR7 agonist) resulted in the significant induction of tube formation in HUVECs and in vivo. Further evidence suggested that CXCL12 induced directional polarization and migration in the HUVECs, which is necessary for tube formation. Moreover, CXCR7 translocalization was observed during the polarization of HUVECs in stripe assays. Finally, treatment with TC14012 also significantly increased PI3K/Akt phosphorylation, and tube formation was blocked by treating HUVECs with an Akt inhibitor. Overall, this study indicated that CXCL12-stimulated CXCR7 acts as a functional receptor to activate Akt for angiogenesis in HUVECs and that CXCR7 may be a potential target molecule for endothelial regeneration and repair after vascular injury.
Collapse
|
40
|
Lu G, He Q, Shen Y, Cao F. Potential biomarkers for predicting hemorrhagic transformation of ischemic stroke. Int J Neurosci 2017; 128:79-89. [PMID: 28726570 DOI: 10.1080/00207454.2017.1349766] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Reperfusion therapy contributes to better clinical outcomes in patients with acute ischemic stroke but carries a more significant risk of hemorrhagic transformation (HT) compared to supportive care. Once HT occurs, the outcome is usually poor and this causes a dilemma in the treatment of ischemic stroke. Consequently, early prediction of HT would be extremely helpful for guiding precise treatment of ischemic stroke. In this review, we focus on summarizing biomarkers of HT and elucidating possible mechanisms so as to identify potential biomarkers for predicting HT.
Collapse
Affiliation(s)
- Guanfeng Lu
- a Department of Neurology, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430022 , China
| | - Quanwei He
- a Department of Neurology, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430022 , China
| | - Yan Shen
- a Department of Neurology, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430022 , China
| | - Fei Cao
- a Department of Neurology, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430022 , China
| |
Collapse
|
41
|
Peña ID, Borlongan C, Shen G, Davis W. Strategies to Extend Thrombolytic Time Window for Ischemic Stroke Treatment: An Unmet Clinical Need. J Stroke 2017; 19:50-60. [PMID: 28178410 PMCID: PMC5307939 DOI: 10.5853/jos.2016.01515] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 01/05/2017] [Accepted: 01/05/2017] [Indexed: 12/13/2022] Open
Abstract
To date, reperfusion with tissue plasminogen activator (tPA) remains the gold standard treatment for ischemic stroke. However, when tPA is given beyond 4.5 hours of stroke onset, deleterious effects of the drug ensue, especially, hemorrhagic transformation (HT), which causes the most significant morbidity and mortality in stroke patients. An important clinical problem at hand is to develop strategies that will enhance the therapeutic time window for tPA therapy and reduce the adverse effects (especially HT) of delayed tPA treatment. We reviewed the pharmacological agents which reduced the risk of HT associated with delayed (beyond 4.5 hours post-stroke) tPA treatment in preclinical studies, which we classified into those that putatively preserve the blood-brain barrier (e.g., minocycline, cilostazol, fasudil, candesartan, and bryostatin) and/or enhance vascularization and protect the cerebrovasculature (e.g., coumarin derivate IMM-H004 and granulocyte colony-stimulating factor). Recently, other new therapeutic modalities (e.g., oxygen transporters) have been reported which improved delayed tPA-associated outcomes by acting through other mechanisms. While the above-mentioned interventions unequivocally reduced delayed tPA-induced HT in stroke models, the long-term efficacy of these drugs are not yet established. Further optimization is required to expedite their future clinical application. The findings from this review indicate the need to explore the most ideal adjunctive interventions that will not only reduce delayed tPA–induced HT, but also preserve neurovascular functions. While waiting for the next breakthrough drug in acute stroke treatment, it is equally important to allocate considerable effort to find approaches to address the limitations of the only FDA-approved stroke therapy.
Collapse
Affiliation(s)
- Ike Dela Peña
- Department of Pharmaceutical and Administrative Sciences, Loma Linda University School of Pharmacy, Loma Linda, United States
| | - Cesar Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, United States
| | - Guofang Shen
- Department of Pharmaceutical and Administrative Sciences, Loma Linda University School of Pharmacy, Loma Linda, United States
| | - Willie Davis
- Department of Pharmaceutical and Administrative Sciences, Loma Linda University School of Pharmacy, Loma Linda, United States
| |
Collapse
|
42
|
England TJ, Sprigg N, Alasheev AM, Belkin AA, Kumar A, Prasad K, Bath PM. Granulocyte-Colony Stimulating Factor (G-CSF) for stroke: an individual patient data meta-analysis. Sci Rep 2016; 6:36567. [PMID: 27845349 PMCID: PMC5109224 DOI: 10.1038/srep36567] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 10/18/2016] [Indexed: 12/11/2022] Open
Abstract
Granulocyte colony stimulating factor (G-CSF) may enhance recovery from stroke through neuroprotective mechanisms if administered early, or neurorepair if given later. Several small trials suggest administration is safe but effects on efficacy are unclear. We searched for randomised controlled trials (RCT) assessing G-CSF in patients with hyperacute, acute, subacute or chronic stroke, and asked Investigators to share individual patient data on baseline characteristics, stroke severity and type, end-of-trial modified Rankin Scale (mRS), Barthel Index, haematological parameters, serious adverse events and death. Multiple variable analyses were adjusted for age, sex, baseline severity and time-to-treatment. Individual patient data were obtained for 6 of 10 RCTs comprising 196 stroke patients (116 G-CSF, 80 placebo), mean age 67.1 (SD 12.9), 92% ischaemic, median NIHSS 10 (IQR 5–15), randomised 11 days (interquartile range IQR 4–238) post ictus; data from three commercial trials were not shared. G-CSF did not improve mRS (ordinal regression), odds ratio OR 1.12 (95% confidence interval 0.64 to 1.96, p = 0.62). There were more patients with a serious adverse event in the G-CSF group (29.6% versus 7.5%, p = 0.07) with no significant difference in all-cause mortality (G-CSF 11.2%, placebo 7.6%, p = 0.4). Overall, G-CSF did not improve stroke outcome in this individual patient data meta-analysis.
Collapse
Affiliation(s)
- Timothy J England
- Vascular Medicine, Division of Medical Sciences and GEM, School of Medicine, University of Nottingham, UK
| | - Nikola Sprigg
- Stroke Trials Unit, Division of Clinical Neuroscience, School of Medicine, University of Nottingham, UK
| | | | - Andrey A Belkin
- Institute of Medical Cell Technologies, Yekaterinburg, Russia
| | - Amit Kumar
- Department of Neurology, Neurosciences Centre, All India Institute of Medical Sciences, New Delhi, India
| | - Kameshwar Prasad
- Department of Neurology, Neurosciences Centre, All India Institute of Medical Sciences, New Delhi, India
| | - Philip M Bath
- Stroke Trials Unit, Division of Clinical Neuroscience, School of Medicine, University of Nottingham, UK
| |
Collapse
|
43
|
Peña ID, Borlongan CV. Translating G-CSF as an Adjunct Therapy to Stem Cell Transplantation for Stroke. Transl Stroke Res 2016; 6:421-9. [PMID: 26482176 DOI: 10.1007/s12975-015-0430-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 10/05/2015] [Accepted: 10/08/2015] [Indexed: 01/26/2023]
Abstract
Among recently investigated stroke therapies, stem cell treatment holds great promise by virtue of their putative ability to replace lost cells, promote endogenous neurogenesis,and produce behavioral and functional improvement through their "bystander effects." Translating stem cell in the clinic, however, presents a number of technical difficulties. A strategy suggested to enhance therapeutic utility of stem cells is combination therapy, i.e., co-transplantation of stem cells or adjunct treatment with pharmacological agents and substrates,which is assumed to produce more profound therapeutic benefits by circumventing limitations of individual treatments and facilitating complementary brain repair processes. We previously demonstrated enhanced functional effects of cotreatment with granulocyte-colony stimulating factor (GCSF)and human umbilical cord blood cell (hUCB) transplantation in animal models of traumatic brain injury (TBI). Here,we suggest that the aforementioned combination therapy may also produce synergistic effects in stroke. Accordingly, G-CSF treatment may reduce expression of pro-inflammatory cytokines and enhance neurogenesis rendering a receptive microenvironment for hUCB engraftment. Adjunct treatment of GCSF with hUCB may facilitate stemness maintenance and guide neural lineage commitment of hUCB cells. Moreover, regenerative mechanisms afforded by G-CSF-mobilized endogenous stem cells, secretion of growth factors by hUCB grafts and G-CSF-recruited endothelial progenitor cells(EPCs), as well as the potential graft–host integration that may promote synaptic circuitry re-establishment could altogether produce more pronounced functional improvement in stroked rats subjected to a combination G-CSF treatment and hUCB transplantation. Nevertheless, differences in pathology and repair processes underlying TBI and stroke deserve consideration when testing the effects of combinatorial G-CSF and hUCB cell transplantation for stroke treatment. Further studies are also required to determine the safety and efficacy of this intervention in both preclinical and clinical stroke studies.
Collapse
|
44
|
Chau M, Zhang J, Wei L, Yu SP. Regeneration after stroke: Stem cell transplantation and trophic factors. Brain Circ 2016; 2:86-94. [PMID: 30276278 PMCID: PMC6126254 DOI: 10.4103/2394-8108.186279] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 06/16/2016] [Accepted: 06/21/2016] [Indexed: 12/13/2022] Open
Abstract
Stroke is a leading cause of death and disability worldwide. However, there is only one Food and Drug Administration-approved drug for the treatment of ischemic stroke, i.e., tissue plasminogen activator, and its therapeutic window is limited to within 4.5 h after stroke. Since clinical trials for neuroprotection have failed to demonstrate efficacy, multipotent and pluripotent stem cell transplantations are viable candidates for stroke treatment by providing trophic factor support and/or cell replacement following injury. The goal of this review is to highlight the promise of stem cell transplantation as vehicles for trophic factor delivery. The beneficial effects of different stem cell types as transplants as well as ways to upregulate trophic factors in stem cells are described in this review. Stem cell transplantation has consistently shown beneficial effects in the ischemic stroke model, in part due to the beneficial factors that stem cells release around the stroke injury area, resulting in smaller infarct volumes and regeneration and functional recovery. Upregulation of beneficial factors in stem cells and neural progenitors before transplantation has been shown to be even more effective in treating the stroke injury than stem cells without upregulated factors. However, for both stem cells and genetic engineering, there remain many unanswered questions and potential for improvement. These include modifiable parameters such as the different stem cell types and different factors, as well as the various readouts for investigation, such as various in vivo effects, such as immune system modulation and enhancement of endogenous neurogenesis and angiogenesis.
Collapse
Affiliation(s)
- Monica Chau
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - James Zhang
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Ling Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Shan Ping Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
45
|
Mizuma A, Yamashita T, Kono S, Nakayama T, Baba Y, Itoh S, Asakura K, Niimi Y, Asahi T, Kanemaru K, Mutoh T, Kuroda S, Kinouchi H, Abe K, Takizawa S. Phase II Trial of Intravenous Low-Dose Granulocyte Colony-Stimulating Factor in Acute Ischemic Stroke. J Stroke Cerebrovasc Dis 2016; 25:1451-7. [DOI: 10.1016/j.jstrokecerebrovasdis.2016.01.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 01/05/2016] [Accepted: 01/11/2016] [Indexed: 01/03/2023] Open
|
46
|
Strecker JK, Olk J, Hoppen M, Gess B, Diederich K, Schmidt A, Schäbitz WR, Schilling M, Minnerup J. Combining Growth Factor and Bone Marrow Cell Therapy Induces Bleeding and Alters Immune Response After Stroke in Mice. Stroke 2016; 47:852-62. [PMID: 26839353 DOI: 10.1161/strokeaha.115.011230] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 12/31/2015] [Indexed: 12/27/2022]
Abstract
BACKGROUND AND PURPOSE Bone marrow cell (BMC)-based therapies, either the transplantation of exogenous cells or stimulation of endogenous cells by growth factors like the granulocyte colony-stimulating factor (G-CSF), are considered a promising means of treating stroke. In contrast to large preclinical evidence, however, a recent clinical stroke trial on G-CSF was neutral. We, therefore, aimed to investigate possible synergistic effects of co-administration of G-CSF and BMCs after experimental stroke in mice to enhance the efficacy compared with single treatments. METHODS We used an animal model for experimental stroke as paradigm to study possible synergistic effects of co-administration of G-CSF and BMCs on the functional outcome and the pathophysiological mechanism. RESULTS G-CSF treatment alone led to an improved functional outcome, a reduced infarct volume, increased blood vessel stabilization, and decreased overall inflammation. Surprisingly, the combination of G-CSF and BMCs abrogated G-CSFs' beneficial effects and resulted in increased hemorrhagic infarct transformation, altered blood-brain barrier, excessive astrogliosis, and altered immune cell polarization. These increased rates of infarct bleeding were mainly mediated by elevated matrix metalloproteinase-9-mediated blood-brain barrier breakdown in G-CSF- and BMCs-treated animals combined with an increased number of dilated and thus likely more fragile vessels in the subacute phase after cerebral ischemia. CONCLUSIONS Our results provide new insights into both BMC-based therapies and immune cell biology and help to understand potential adverse and unexpected side effects.
Collapse
Affiliation(s)
- Jan-Kolja Strecker
- From the Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, Münster, Germany (J.-K.S., J.O., M.H., B.G., K.D., A.S., M.S., J.M.); and EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.).
| | - Joanna Olk
- From the Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, Münster, Germany (J.-K.S., J.O., M.H., B.G., K.D., A.S., M.S., J.M.); and EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.)
| | - Maike Hoppen
- From the Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, Münster, Germany (J.-K.S., J.O., M.H., B.G., K.D., A.S., M.S., J.M.); and EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.)
| | - Burkhard Gess
- From the Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, Münster, Germany (J.-K.S., J.O., M.H., B.G., K.D., A.S., M.S., J.M.); and EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.)
| | - Kai Diederich
- From the Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, Münster, Germany (J.-K.S., J.O., M.H., B.G., K.D., A.S., M.S., J.M.); and EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.)
| | - Antje Schmidt
- From the Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, Münster, Germany (J.-K.S., J.O., M.H., B.G., K.D., A.S., M.S., J.M.); and EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.)
| | - Wolf-Rüdiger Schäbitz
- From the Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, Münster, Germany (J.-K.S., J.O., M.H., B.G., K.D., A.S., M.S., J.M.); and EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.)
| | - Matthias Schilling
- From the Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, Münster, Germany (J.-K.S., J.O., M.H., B.G., K.D., A.S., M.S., J.M.); and EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.)
| | - Jens Minnerup
- From the Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, Münster, Germany (J.-K.S., J.O., M.H., B.G., K.D., A.S., M.S., J.M.); and EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.)
| |
Collapse
|
47
|
Henninger N, Fisher M. Extending the Time Window for Endovascular and Pharmacological Reperfusion. Transl Stroke Res 2016; 7:284-93. [DOI: 10.1007/s12975-015-0444-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 12/13/2015] [Accepted: 12/14/2015] [Indexed: 01/07/2023]
|
48
|
Wang W, Li M, Wang Y, Li Q, Deng G, Wan J, Yang Q, Chen Q, Wang J. GSK-3β inhibitor TWS119 attenuates rtPA-induced hemorrhagic transformation and activates the Wnt/β-catenin signaling pathway after acute ischemic stroke in rats. Mol Neurobiol 2015; 53:7028-7036. [PMID: 26671619 DOI: 10.1007/s12035-015-9607-2] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Accepted: 12/03/2015] [Indexed: 10/22/2022]
Abstract
Hemorrhagic transformation (HT) is a devastating complication for patients with acute ischemic stroke who are treated with tissue plasminogen activator (tPA). It is associated with high morbidity and mortality, but no effective treatments are currently available to reduce HT risk. Therefore, methods to prevent HT are urgently needed. In this study, we used TWS119, an inhibitor of glycogen synthase kinase 3β (GSK-3β), to evaluate the role of the Wnt/β-catenin signaling pathway in recombinant tPA (rtPA)-induced HT. Sprague-Dawley rats were subjected to a middle cerebral artery occlusion (MCAO) model of ischemic stroke and then were administered rtPA, rtPA combined with TWS119, or vehicle at 4 h. The animals were sacrificed 24 h after infarct induction. Rats treated with rtPA showed evident HT, had more severe neurologic deficit, brain edema, and blood-brain barrier breakdown, and had larger infarction volume than did the vehicle group. Rats treated with TWS119 had significantly improved outcomes compared with those of rats treated with rtPA alone. In addition, Western blot analysis showed that TWS119 increased the protein expression of β-catenin, claudin-3, and ZO-1 while suppressing the expression of GSK-3β. These results suggest that TWS119 reduces rtPA-induced HT and attenuates blood-brain barrier disruption, possibly through activation of the Wnt/β-catenin signaling pathway. This study provides a potential therapeutic strategy to prevent tPA-induced HT after acute ischemic stroke.
Collapse
Affiliation(s)
- Wei Wang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Mingchang Li
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yuefei Wang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Qian Li
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, 720 Rutland Ave, Ross Bldg 370B, Baltimore, MD, 21205, USA
| | - Gang Deng
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jieru Wan
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, 720 Rutland Ave, Ross Bldg 370B, Baltimore, MD, 21205, USA
| | - Qingwu Yang
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400044, China
| | - Qianxue Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Jian Wang
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, 720 Rutland Ave, Ross Bldg 370B, Baltimore, MD, 21205, USA.
| |
Collapse
|
49
|
Wallner S, Peters S, Pitzer C, Resch H, Bogdahn U, Schneider A. The Granulocyte-colony stimulating factor has a dual role in neuronal and vascular plasticity. Front Cell Dev Biol 2015; 3:48. [PMID: 26301221 PMCID: PMC4528279 DOI: 10.3389/fcell.2015.00048] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Accepted: 07/23/2015] [Indexed: 12/13/2022] Open
Abstract
Granulocyte-colony stimulating factor (G-CSF) is a growth factor that has originally been identified several decades ago as a hematopoietic factor required mainly for the generation of neutrophilic granulocytes, and is in clinical use for that. More recently, it has been discovered that G-CSF also plays a role in the brain as a growth factor for neurons and neural stem cells, and as a factor involved in the plasticity of the vasculature. We review and discuss these dual properties in view of the neuroregenerative potential of this growth factor.
Collapse
Affiliation(s)
- Stephanie Wallner
- Department of Traumatology and Sports Injuries, Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University SalzburgSalzburg, Austria
| | - Sebastian Peters
- Department of Neurology, University Hospital RegensburgRegensburg, Germany
| | - Claudia Pitzer
- Interdisciplinary Neurobehavioral Core, Ruprecht-Karls-UniversityHeidelberg, Germany
| | - Herbert Resch
- Department of Traumatology and Sports Injuries, Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University SalzburgSalzburg, Austria
- University Clinic of Traumatology and Sports Injuries Salzburg, Paracelsus Medical University SalzburgSalzburg, Austria
| | - Ulrich Bogdahn
- Department of Neurology, University Hospital RegensburgRegensburg, Germany
| | | |
Collapse
|