101
|
Szeto V, Chen NH, Sun HS, Feng ZP. The role of K ATP channels in cerebral ischemic stroke and diabetes. Acta Pharmacol Sin 2018; 39:683-694. [PMID: 29671418 PMCID: PMC5943906 DOI: 10.1038/aps.2018.10] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 02/19/2018] [Indexed: 12/18/2022] Open
Abstract
ATP-sensitive potassium (KATP) channels are ubiquitously expressed on the plasma membrane of cells in multiple organs, including the heart, pancreas and brain. KATP channels play important roles in controlling and regulating cellular functions in response to metabolic state, which are inhibited by ATP and activated by Mg-ADP, allowing the cell to couple cellular metabolic state (ATP/ADP ratio) to electrical activity of the cell membrane. KATP channels mediate insulin secretion in pancreatic islet beta cells, and controlling vascular tone. Under pathophysiological conditions, KATP channels play cytoprotective role in cardiac myocytes and neurons during ischemia and/or hypoxia. KATP channel is a hetero-octameric complex, consisting of four pore-forming Kir6.x and four regulatory sulfonylurea receptor SURx subunits. These subunits are differentially expressed in various cell types, thus determining the sensitivity of the cells to specific channel modifiers. Sulfonylurea class of antidiabetic drugs blocks KATP channels, which are neuroprotective in stroke, can be one of the high stoke risk factors for diabetic patients. In this review, we discussed the potential effects of KATP channel blockers when used under pathological conditions related to diabetics and cerebral ischemic stroke.
Collapse
Affiliation(s)
- Vivian Szeto
- Departments of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada M5S 1A8
| | - Nai-hong Chen
- Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Hong-shuo Sun
- Departments of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada M5S 1A8
- Surgery
- Pharmacology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada M5S 1A8
| | - Zhong-ping Feng
- Departments of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada M5S 1A8
| |
Collapse
|
102
|
Bylicky MA, Mueller GP, Day RM. Mechanisms of Endogenous Neuroprotective Effects of Astrocytes in Brain Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:6501031. [PMID: 29805731 PMCID: PMC5901819 DOI: 10.1155/2018/6501031] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 02/19/2018] [Indexed: 12/11/2022]
Abstract
Astrocytes, once believed to serve only as "glue" for the structural support of neurons, have been demonstrated to serve critical functions for the maintenance and protection of neurons, especially under conditions of acute or chronic injury. There are at least seven distinct mechanisms by which astrocytes protect neurons from damage; these are (1) protection against glutamate toxicity, (2) protection against redox stress, (3) mediation of mitochondrial repair mechanisms, (4) protection against glucose-induced metabolic stress, (5) protection against iron toxicity, (6) modulation of the immune response in the brain, and (7) maintenance of tissue homeostasis in the presence of DNA damage. Astrocytes support these critical functions through specialized responses to stress or toxic conditions. The detoxifying activities of astrocytes are essential for maintenance of the microenvironment surrounding neurons and in whole tissue homeostasis. Improved understanding of the mechanisms by which astrocytes protect the brain could lead to the development of novel targets for the development of neuroprotective strategies.
Collapse
Affiliation(s)
- Michelle A. Bylicky
- Department of Anatomy, Physiology, and Genetics, The Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Gregory P. Mueller
- Department of Anatomy, Physiology, and Genetics, The Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Regina M. Day
- Department of Pharmacology and Molecular Therapeutics, The Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| |
Collapse
|
103
|
Zhao X, Zhou K, Li Z, Nan W, Wang J, Xia Y, Zhang H. Knockdown of Ski decreased the reactive astrocytes proliferation in vitro induced by oxygen‐glucose deprivation/reoxygenation. J Cell Biochem 2018; 119:4548-4558. [DOI: 10.1002/jcb.26597] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 12/07/2017] [Indexed: 12/17/2022]
Affiliation(s)
- Xin Zhao
- The Second Clinical Medical College of Lanzhou UniversityLanzhouPR China
- Orthopaedics Key laboratory of Gansu ProvinceLanzhouPR China
| | - Kai‐Sheng Zhou
- The Second Clinical Medical College of Lanzhou UniversityLanzhouPR China
- Orthopaedics Key laboratory of Gansu ProvinceLanzhouPR China
| | - Zhong‐Hao Li
- The Second Clinical Medical College of Lanzhou UniversityLanzhouPR China
- Orthopaedics Key laboratory of Gansu ProvinceLanzhouPR China
| | - Wei Nan
- The Second Clinical Medical College of Lanzhou UniversityLanzhouPR China
- Orthopaedics Key laboratory of Gansu ProvinceLanzhouPR China
| | - Jing Wang
- Orthopaedics Key laboratory of Gansu ProvinceLanzhouPR China
| | - Ya‐Yi Xia
- The Second Clinical Medical College of Lanzhou UniversityLanzhouPR China
| | - Hai‐Hong Zhang
- The Second Clinical Medical College of Lanzhou UniversityLanzhouPR China
| |
Collapse
|
104
|
Takarada-Iemata M, Yoshikawa A, Ta HM, Okitani N, Nishiuchi T, Aida Y, Kamide T, Hattori T, Ishii H, Tamatani T, Le TM, Roboon J, Kitao Y, Matsuyama T, Nakada M, Hori O. N-myc downstream-regulated gene 2 protects blood-brain barrier integrity following cerebral ischemia. Glia 2018; 66:1432-1446. [PMID: 29476556 DOI: 10.1002/glia.23315] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 12/22/2017] [Accepted: 02/09/2018] [Indexed: 11/10/2022]
Abstract
Disruption of the blood-brain barrier (BBB) following cerebral ischemia is closely related to the infiltration of peripheral cells into the brain, progression of lesion formation, and clinical exacerbation. However, the mechanism that regulates BBB integrity, especially after permanent ischemia, remains unclear. Here, we present evidence that astrocytic N-myc downstream-regulated gene 2 (NDRG2), a differentiation- and stress-associated molecule, may function as a modulator of BBB permeability following ischemic stroke, using a mouse model of permanent cerebral ischemia. Immunohistological analysis showed that the expression of NDRG2 increases dominantly in astrocytes following permanent middle cerebral artery occlusion (MCAO). Genetic deletion of Ndrg2 exhibited enhanced levels of infarct volume and accumulation of immune cells into the ipsilateral brain hemisphere following ischemia. Extravasation of serum proteins including fibrinogen and immunoglobulin, after MCAO, was enhanced at the ischemic core and perivascular region of the peri-infarct area in the ipsilateral cortex of Ndrg2-deficient mice. Furthermore, the expression of matrix metalloproteinases (MMPs) after MCAO markedly increased in Ndrg2-/- mice. In culture, expression and secretion of MMP-3 was increased in Ndrg2-/- astrocytes, and this increase was reversed by adenovirus-mediated re-expression of NDRG2. These findings suggest that NDRG2, expressed in astrocytes, may play a critical role in the regulation of BBB permeability and immune cell infiltration through the modulation of MMP expression following cerebral ischemia.
Collapse
Affiliation(s)
- Mika Takarada-Iemata
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Akifumi Yoshikawa
- Department of Neurosurgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, Ishikawa, 920-8641, Japan
| | - Hieu Minh Ta
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Nahoko Okitani
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Takumi Nishiuchi
- Division of Functional Genomics, Advanced Science Research Center, Kanazawa University, 13-1 Takara-Machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Yasuhiro Aida
- Department of Neurosurgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, Ishikawa, 920-8641, Japan
| | - Tomoya Kamide
- Department of Neurosurgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, Ishikawa, 920-8641, Japan
| | - Tsuyoshi Hattori
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Hiroshi Ishii
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Takashi Tamatani
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Thuong Manh Le
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Jureepon Roboon
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Yasuko Kitao
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Tomohiro Matsuyama
- Laboratory of Neurogenesis and CNS Repair, Institute for Advanced Medical Sciences, Hyogo College of Medicine, 1-1 Mukogawa-Machi, Nishinomiya, Hyogo, 663-8501, Japan
| | - Mitsutoshi Nakada
- Department of Neurosurgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, Ishikawa, 920-8641, Japan
| | - Osamu Hori
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, Ishikawa, 920-8640, Japan
| |
Collapse
|
105
|
Ras-Related C3 Botulinum Toxin Substrate 1 Promotes Axonal Regeneration after Stroke in Mice. Transl Stroke Res 2018; 9:506-514. [PMID: 29476448 DOI: 10.1007/s12975-018-0611-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 01/09/2018] [Accepted: 01/10/2018] [Indexed: 12/18/2022]
Abstract
Neurite plasticity is a critical aspect of brain functional recovery after stroke. Emerging data suggest that Ras-related C3 botulinum toxin substrate 1 (Rac1) plays a central role in axonal regeneration in the injured brain, specifically by stimulating neuronal intrinsic growth and counteracting the growth inhibitory signaling that leads to growth cone collapse. Therefore, we investigated the functional role of Rac1 in axonal regeneration after stroke.Delayed treatment with a specific Rac1 inhibitor, NSC 23766, worsened functional recovery, which was assessed by the pellet reaching test from day 14 to day 28 after stroke. It additionally reduced axonal density in the peri-infarct zone, assessed 28 days after stroke, with no effect on brain cavity size or on the number of newly formed cells. Accordingly, Rac1 overexpression using lentivirus promoted axonal regeneration and functional recovery after stroke from day 14 to day 28. Rac1 inhibition led to inactivation of pro-regenerative molecules, including mitogen-activated protein kinase kinase (p-MEK)1/2, LIM domain kinase (LIMK)1, and extracellular signal-regulated kinase (p-ERK)1/2 at 14 days after stroke. Inhibition of Rac1 reduced axonal length and number in cultured primary mouse cortical neurons using microfluidic chambers after oxygen-glucose deprivation (OGD) without affecting cell viability. In contrast, inhibition of Rac1 increased levels of glial fibrillary acidic protein, an extrinsic inhibitory signal for axonal growth, after stroke in vivo and in primary astrocytes after OGD.In conclusion, Rac1 signaling enhances axonal regeneration and improve post-stroke functional recovery in experimental models of stroke.
Collapse
|
106
|
Focal Ischaemic Infarcts Expand Faster in Cerebellar Cortex than Cerebral Cortex in a Mouse Photothrombotic Stroke Model. Transl Stroke Res 2018; 9:643-653. [PMID: 29455391 DOI: 10.1007/s12975-018-0615-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 02/02/2018] [Accepted: 02/04/2018] [Indexed: 12/31/2022]
Abstract
It is generally accepted that the cerebellum is particularly vulnerable to ischaemic injury, and this may contribute to the high mortality arising from posterior circulation strokes. However, this has not been systematically examined in an animal model. This study compared the development and resolution of matched photothrombotic microvascular infarcts in the cerebellar and cerebral cortices in adult 129/SvEv mice of both sexes. The photothrombotic lesions were made using tail vein injection of Rose Bengal with a 532 nm laser projected onto a 2 mm diameter aperture over the target region of the brain (with skull thinning). Infarct size was then imaged histologically following 2 h to 30-day survival using serial reconstruction of haematoxylin and eosin stained cryosections. This was complemented with immunohistochemistry for neuron and glial markers. At 2 h post-injury, the cerebellar infarct volume averaged ~ 2.7 times that of the cerebral cortex infarcts. Infarct volume reached maximum in the cerebellum in a quarter of the time (24 h) taken in the cerebral cortex (4 days). Remodelling resolved the infarcts within a month, leaving significantly larger residual injury volume in the cerebellum. The death of neurons in the core lesion at 2 h was confirmed by NeuN and Calbindin immunofluorescence, alongside activation of astrocytes and microglia. The latter persisted in the region within and surrounding the residual infarct at 30 days. This comparison of acute focal ischaemic injuries in cerebellar and cerebral cortices provides direct confirmation of exacerbation of neuropathology and faster kinetics in the cerebellum.
Collapse
|
107
|
Thompson R, Sakiyama-Elbert S. Using biomaterials to promote pro-regenerative glial phenotypes after nervous system injuries. ACTA ACUST UNITED AC 2018; 13:024104. [PMID: 29186011 DOI: 10.1088/1748-605x/aa9e23] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Trauma to either the central or peripheral nervous system (PNS) often leads to significant loss of function and disability in patients. This high rate of long-term disability is due to the overall limited regenerative potential of nervous tissue, even though the PNS has more regenerative potential than the central nervous system (CNS). The supporting glial cells in the periphery, Schwann cells, are part of the reason for the improved recovery observed in the PNS. In the CNS, the glial populations, astrocytes and oligodendrocytes (OLs), do not have as much potential to promote regeneration and are at times inhibitory to neuronal growth. In particular, the inhibitory roles astrocytes play following trauma has led to a historical focus on neurons and OLs instead of astrocytes. Recently, this focus has shifted as new, regenerative astrocyte phenotypes have been described. From these observations, glial cells clearly play critical roles in native recovery pathways in both the CNS and PNS. This makes the ability to manipulate both transplanted and native glial cell phenotypes a potentially successful strategy to improve nerve injury outcomes. This review focuses on factors that cause glial cells to adopt repair phenotypes and biomaterials that manipulate and/or harness these glial phenotypes.
Collapse
Affiliation(s)
- Russell Thompson
- Department of Biomedical Engineering, University of Texas at Austin 107 W Dean Keeton, Austin, TX 78712, United States of America. Department of Biomedical Engineering, Washington University in St. Louis, 1 Brooking Drive, St. Louis, MO 63130, United States of America
| | | |
Collapse
|
108
|
Laterza C, Uoshima N, Tornero D, Wilhelmsson U, Stokowska A, Ge R, Pekny M, Lindvall O, Kokaia Z. Attenuation of reactive gliosis in stroke-injured mouse brain does not affect neurogenesis from grafted human iPSC-derived neural progenitors. PLoS One 2018; 13:e0192118. [PMID: 29401502 PMCID: PMC5798785 DOI: 10.1371/journal.pone.0192118] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 01/18/2018] [Indexed: 11/19/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) or their progeny, derived from human somatic cells, can give rise to functional improvements after intracerebral transplantation in animal models of stroke. Previous studies have indicated that reactive gliosis, which is associated with stroke, inhibits neurogenesis from both endogenous and grafted neural stem/progenitor cells (NSPCs) of rodent origin. Here we have assessed whether reactive astrocytes affect the fate of human iPSC-derived NSPCs transplanted into stroke-injured brain. Mice with genetically attenuated reactive gliosis (deficient for GFAP and vimentin) were subjected to cortical stroke and cells were implanted adjacent to the ischemic lesion one week later. At 8 weeks after transplantation, immunohistochemical analysis showed that attenuated reactive gliosis did not affect neurogenesis or commitment towards glial lineage of the grafted NSPCs. Our findings, obtained in a human-to-mouse xenograft experiment, provide evidence that the reactive gliosis in stroke-injured brain does not affect the formation of new neurons from intracortically grafted human iPSC-derived NSPCs. However, for a potential clinical translation of these cells in stroke, it will be important to clarify whether the lack of effect of reactive gliosis on neurogenesis is observed also in a human-to-human experimental setting.
Collapse
Affiliation(s)
- Cecilia Laterza
- Department of Clinical Sciences, Laboratory of Stem Cells & Restorative Neurology, Lund Stem Cell Center, University Hospital, Lund, Sweden
| | - Naomi Uoshima
- Department of Clinical Sciences, Laboratory of Stem Cells & Restorative Neurology, Lund Stem Cell Center, University Hospital, Lund, Sweden
- Department of Anesthesiology, Tokyo Medical University, Nishishinjuku, Shinjuku-ku, Tokyo, Japan
| | - Daniel Tornero
- Department of Clinical Sciences, Laboratory of Stem Cells & Restorative Neurology, Lund Stem Cell Center, University Hospital, Lund, Sweden
| | - Ulrika Wilhelmsson
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Laboratory of Astrocyte Biology and CNS Regeneration, Center for Brain Repair, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Anna Stokowska
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Laboratory of Regenerative Neuroimmunology, Center for Brain Repair, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Ruimin Ge
- Department of Clinical Sciences, Laboratory of Stem Cells & Restorative Neurology, Lund Stem Cell Center, University Hospital, Lund, Sweden
| | - Milos Pekny
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Laboratory of Astrocyte Biology and CNS Regeneration, Center for Brain Repair, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Olle Lindvall
- Department of Clinical Sciences, Laboratory of Stem Cells & Restorative Neurology, Lund Stem Cell Center, University Hospital, Lund, Sweden
| | - Zaal Kokaia
- Department of Clinical Sciences, Laboratory of Stem Cells & Restorative Neurology, Lund Stem Cell Center, University Hospital, Lund, Sweden
- * E-mail:
| |
Collapse
|
109
|
Alaverdashvili M, Caine S, Li X, Hackett MJ, Bradley MP, Nichol H, Paterson PG. Protein-Energy Malnutrition Exacerbates Stroke-Induced Forelimb Abnormalities and Dampens Neuroinflammation. Transl Stroke Res 2018; 9:622-630. [PMID: 29397529 DOI: 10.1007/s12975-018-0613-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 01/20/2018] [Accepted: 01/23/2018] [Indexed: 11/30/2022]
Abstract
Protein-energy malnutrition (PEM) pre-existing at stroke onset is believed to worsen functional outcome, yet the underlying mechanisms are not fully understood. Since brain inflammation is an important modulator of neurological recovery after stroke, we explored the impact of PEM on neuroinflammation in the acute period in relation to stroke-initiated sensori-motor abnormalities. Adult rats were fed a low-protein (LP) or normal protein (NP) diet for 28 days before inducing photothrombotic stroke (St) in the forelimb region of the motor cortex or sham surgery; the diets continued for 3 days after the stroke. Protein-energy status was assessed by a combination of body weight, food intake, serum acute phase proteins and corticosterone, and liver lipid content. Deficits in motor function were evaluated in the horizontal ladder walking and cylinder tasks at 3 days after stroke. The glial response and brain elemental signature were investigated by immunohistochemistry and micro-X-ray fluorescence imaging, respectively. The LP-fed rats reduced food intake, resulting in PEM. Pre-existing PEM augmented stroke-induced abnormalities in forelimb placement accuracy on the ladder; LP-St rats made more errors (29 ± 8%) than the NP-St rats (15 ± 3%; P < 0.05). This was accompanied by attenuated astrogliosis in the peri-infarct area by 18% and reduced microglia activation by up to 41 and 21% in the peri-infarct area and the infarct rim, respectively (P < 0.05). The LP diet altered the cortical Zn, Ca, and Cl signatures (P < 0.05). Our data suggest that proactive treatment of pre-existing PEM could be essential for optimal post-stroke recovery.
Collapse
Affiliation(s)
- Mariam Alaverdashvili
- College of Pharmacy and Nutrition, University of Saskatchewan, D Wing Health Sciences, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada.
| | - Sally Caine
- College of Pharmacy and Nutrition, University of Saskatchewan, D Wing Health Sciences, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada
| | - Xue Li
- College of Pharmacy and Nutrition, University of Saskatchewan, D Wing Health Sciences, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada
| | - Mark J Hackett
- Department of Geological Sciences, University of Saskatchewan, Saskatoon, Canada
| | - Michael P Bradley
- Department of Physics and Engineering Physics, University of Saskatchewan, Saskatoon, Canada
| | - Helen Nichol
- Department of Anatomy and Cell Biology, University of Saskatchewan, Saskatoon, Canada
| | - Phyllis G Paterson
- College of Pharmacy and Nutrition, University of Saskatchewan, D Wing Health Sciences, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada.
| |
Collapse
|
110
|
RGMa mediates reactive astrogliosis and glial scar formation through TGFβ1/Smad2/3 signaling after stroke. Cell Death Differ 2018; 25:1503-1516. [PMID: 29396549 PMCID: PMC6113216 DOI: 10.1038/s41418-018-0058-y] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 12/22/2017] [Accepted: 12/27/2017] [Indexed: 01/11/2023] Open
Abstract
In response to stroke, astrocytes become reactive astrogliosis and are a major component of a glial scar. This results in the formation of both a physical and chemical (production of chondroitin sulfate proteoglycans) barrier, which prevent neurite regeneration that, in turn, interferes with functional recovery. However, the mechanisms of reactive astrogliosis and glial scar formation are poorly understood. In this work, we hypothesized that repulsive guidance molecule a (RGMa) regulate reactive astrogliosis and glial scar formation. We first found that RGMa was strongly expressed by reactive astrocytes in the glial scar in a rat model of middle cerebral artery occlusion/reperfusion. Genetic or pharmacologic inhibition of RGMa in vivo resulted in a strong reduction of reactive astrogliosis and glial scarring as well as in a pronounced improvement in functional recovery. Furthermore, we showed that transforming growth factor β1 (TGFβ1) stimulated RGMa expression through TGFβ1 receptor activin-like kinase 5 (ALK5) in primary cultured astrocytes. Knockdown of RGMa abrogated key steps of reactive astrogliosis and glial scar formation induced by TGFβ1, including cellular hypertrophy, glial fibrillary acidic protein upregulation, cell migration, and CSPGs secretion. Finally, we demonstrated that RGMa co-immunoprecipitated with ALK5 and Smad2/3. TGFβ1-induced ALK5-Smad2/3 interaction and subsequent phosphorylation of Smad2/3 were impaired by RGMa knockdown. Taken together, we identified that after stroke, RGMa promotes reactive astrogliosis and glial scar formation by forming a complex with ALK5 and Smad2/3 to promote ALK5-Smad2/3 interaction to facilitate TGFβ1/Smad2/3 signaling, thereby inhibiting neurological functional recovery. RGMa may be a new therapeutic target for stroke.
Collapse
|
111
|
Neves JD, Mestriner RG, Netto CA. Astrocytes in the cerebral cortex play a role in the spontaneous motor recovery following experimental striatal hemorrhage. Neural Regen Res 2018; 13:67-68. [PMID: 29451208 PMCID: PMC5840993 DOI: 10.4103/1673-5374.224372] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Juliana Dalibor Neves
- Graduate Program of Physiology, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul; Department of Biochemistry, Institute of Basic Health Science, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Régis Gemerasca Mestriner
- Neurorehabilitation and Neural Repair Research Group, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Carlos Alexandre Netto
- Graduate Program of Physiology, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul; Department of Biochemistry, Institute of Basic Health Science, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
112
|
Alamri FF, Shoyaib AA, Biggers A, Jayaraman S, Guindon J, Karamyan VT. Applicability of the grip strength and automated von Frey tactile sensitivity tests in the mouse photothrombotic model of stroke. Behav Brain Res 2018; 336:250-255. [DOI: 10.1016/j.bbr.2017.09.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 08/29/2017] [Accepted: 09/04/2017] [Indexed: 12/19/2022]
|
113
|
Jha MK, Kim JH, Song GJ, Lee WH, Lee IK, Lee HW, An SSA, Kim S, Suk K. Functional dissection of astrocyte-secreted proteins: Implications in brain health and diseases. Prog Neurobiol 2017; 162:37-69. [PMID: 29247683 DOI: 10.1016/j.pneurobio.2017.12.003] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 10/23/2017] [Accepted: 12/08/2017] [Indexed: 02/07/2023]
Abstract
Astrocytes, which are homeostatic cells of the central nervous system (CNS), display remarkable heterogeneity in their morphology and function. Besides their physical and metabolic support to neurons, astrocytes modulate the blood-brain barrier, regulate CNS synaptogenesis, guide axon pathfinding, maintain brain homeostasis, affect neuronal development and plasticity, and contribute to diverse neuropathologies via secreted proteins. The identification of astrocytic proteome and secretome profiles has provided new insights into the maintenance of neuronal health and survival, the pathogenesis of brain injury, and neurodegeneration. Recent advances in proteomics research have provided an excellent catalog of astrocyte-secreted proteins. This review categorizes astrocyte-secreted proteins and discusses evidence that astrocytes play a crucial role in neuronal activity and brain function. An in-depth understanding of astrocyte-secreted proteins and their pathways is pivotal for the development of novel strategies for restoring brain homeostasis, limiting brain injury/inflammation, counteracting neurodegeneration, and obtaining functional recovery.
Collapse
Affiliation(s)
- Mithilesh Kumar Jha
- Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jong-Heon Kim
- Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Gyun Jee Song
- Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Won-Ha Lee
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - In-Kyu Lee
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Ho-Won Lee
- Department of Neurology, Brain Science and Engineering Institute, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Seong Soo A An
- Department of BioNano Technology, Gachon University, Gyeonggi-do, Republic of Korea
| | - SangYun Kim
- Department of Neurology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Gyeonggi-do, Republic of Korea
| | - Kyoungho Suk
- Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea.
| |
Collapse
|
114
|
Lee HI, Lee SW, Kim NG, Park KJ, Choi BT, Shin YI, Shin HK. Low-level light emitting diode therapy promotes long-term functional recovery after experimental stroke in mice. JOURNAL OF BIOPHOTONICS 2017; 10:1761-1771. [PMID: 28464523 DOI: 10.1002/jbio.201700038] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 03/06/2017] [Accepted: 03/10/2017] [Indexed: 06/07/2023]
Abstract
We aimed to investigate the effects of low-level light emitting diode therapy (LED-T) on the long-term functional outcomes after cerebral ischemia, and the optimal timing of LED-T initiation for achieving suitable functional recovery. Focal cerebral ischemia was induced in mice via photothrombosis. These mice were assigned to a sham-operated (control), ischemic (vehicle), or LED-T group [initiation immediately (acute), 4 days (subacute) or 10 days (delayed) after ischemia, followed by once-daily treatment for 7 days]. Behavioral outcomes were assessed 21 and 28 days post-ischemia, and histopathological analysis was performed 28 days post-ischemia. The acute and subacute LED-T groups showed a significant improvement in motor function up to 28 days post-ischemia, although no brain atrophy recovery was noted. We observed proliferating cells (BrdU+ ) in the ischemic brain, and significant increases in BrdU+ /GFAP+ , BrdU+ /DCX+ , BrdU+ /NeuN+ , and CD31+ cells in the subacute LED-T group. However, the BrdU+ /Iba-1+ cell count was reduced in the subacute LED-T group. Furthermore, the brain-derived neurotrophic factor (BDNF) was significantly upregulated in the subacute LED-T group. We concluded that LED-T administered during the subacute stage had a positive impact on the long-term functional outcome, probably via neuron and astrocyte proliferation, blood vessel reconstruction, and increased BDNF expression. Picture: The rotarod test for motor coordination showed that acute and subacute LED-T improves long-term functional recovery after cerebral ischemia.
Collapse
Affiliation(s)
- Hae In Lee
- Department of Rehabilitation Medicine, School of Medicine, Pusan National University, Yangsan, Gyeongnam, 50612, Republic of Korea
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Gyeongnam, 50612, Republic of Korea
| | - Sae-Won Lee
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam, 50612, Republic of Korea
- Graduate Training Program of Korean Medicine for Healthy-Aging, Pusan National University, Yangsan, Gyeongnam, 50612, Republic of Korea
- Korean Medical Science Research Center for Healthy-Aging, Pusan National University, Yangsan, Gyeongnam, 50612, Republic of Korea
| | - Nam Gyun Kim
- Medical Research Center of Color Seven, Seoul, 06719, Republic of Korea
| | - Kyoung-Jun Park
- Medical Research Center of Color Seven, Seoul, 06719, Republic of Korea
| | - Byung Tae Choi
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam, 50612, Republic of Korea
- Graduate Training Program of Korean Medicine for Healthy-Aging, Pusan National University, Yangsan, Gyeongnam, 50612, Republic of Korea
- Korean Medical Science Research Center for Healthy-Aging, Pusan National University, Yangsan, Gyeongnam, 50612, Republic of Korea
| | - Yong-Il Shin
- Department of Rehabilitation Medicine, School of Medicine, Pusan National University, Yangsan, Gyeongnam, 50612, Republic of Korea
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Gyeongnam, 50612, Republic of Korea
| | - Hwa Kyoung Shin
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam, 50612, Republic of Korea
- Graduate Training Program of Korean Medicine for Healthy-Aging, Pusan National University, Yangsan, Gyeongnam, 50612, Republic of Korea
- Korean Medical Science Research Center for Healthy-Aging, Pusan National University, Yangsan, Gyeongnam, 50612, Republic of Korea
| |
Collapse
|
115
|
Luo H, Wu XQ, Zhao M, Wang Q, Jiang GP, Cai WJ, Luo MY. Expression of vimentin and glial fibrillary acidic protein in central nervous system development of rats. ASIAN PAC J TROP MED 2017; 10:1185-1189. [DOI: 10.1016/j.apjtm.2017.10.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 09/26/2017] [Accepted: 10/25/2017] [Indexed: 11/24/2022] Open
|
116
|
Schönfeld LM, Dooley D, Jahanshahi A, Temel Y, Hendrix S. Evaluating rodent motor functions: Which tests to choose? Neurosci Biobehav Rev 2017; 83:298-312. [PMID: 29107829 DOI: 10.1016/j.neubiorev.2017.10.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 09/18/2017] [Accepted: 10/23/2017] [Indexed: 01/11/2023]
Abstract
Damage to the motor cortex induced by stroke or traumatic brain injury (TBI) can result in chronic motor deficits. For the development and improvement of therapies, animal models which possess symptoms comparable to the clinical population are used. However, the use of experimental animals raises valid ethical and methodological concerns. To decrease discomfort by experimental procedures and to increase the quality of results, non-invasive and sensitive rodent motor tests are needed. A broad variety of rodent motor tests are available to determine deficits after stroke or TBI. The current review describes and evaluates motor tests that fall into three categories: Tests to evaluate fine motor skills and grip strength, tests for gait and inter-limb coordination and neurological deficit scores. In this review, we share our thoughts on standardized data presentation to increase data comparability between studies. We also critically evaluate current methods and provide recommendations for choosing the best behavioral test for a new research line.
Collapse
Affiliation(s)
- Lisa-Maria Schönfeld
- Comparative Psychology, Institute of Experimental Psychology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany.
| | - Dearbhaile Dooley
- Health Science Centre, School of Medicine, University College Dublin, Belfield, Dublin, Ireland
| | - Ali Jahanshahi
- Department of Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Yasin Temel
- Department of Neuroscience, Maastricht University, Maastricht, the Netherlands; Department of Neurosurgery, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Sven Hendrix
- Department of Morphology, Biomedical Research Institute (BIOMED), Hasselt University, Hasselt, Belgium.
| |
Collapse
|
117
|
van der Meulen PM, Barendregt AM, Cuadrado E, Magro-Checa C, Steup-Beekman GM, Schonenberg-Meinema D, Van den Berg JM, Li QZ, Baars PA, Wouters D, Voskuyl AE, Ten Berge IRJM, Huizinga TWJ, Kuijpers TW. Protein array autoantibody profiles to determine diagnostic markers for neuropsychiatric systemic lupus erythematosus. Rheumatology (Oxford) 2017; 56:1407-1416. [PMID: 28460084 DOI: 10.1093/rheumatology/kex073] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Indexed: 12/11/2022] Open
Abstract
Objective The aim was to investigate the association between autoantibodies (autoAbs) and neuropsychiatric (NP) involvement in patients with SLE and to evaluate whether any autoAb or a combination of these autoAbs could indicate the underlying pathogenic process. Methods Using a multiplexed protein array for 94 antigens, we compared the serum autoAb profiles of 69 NPSLE patients, 203 SLE patients without NP involvement (non-NPSLE) and 51 healthy controls. Furthermore, we compared the profiles of NPSLE patients with clinical inflammatory (n = 38) and ischaemic (n = 31) NP involvement. Results In total, 75 IgG and 47 IgM autoAbs were associated with SLE patients in comparison with healthy controls. Comparing NPSLE with non-NPSLE and healthy control sera, 9 IgG (amyloid, cardiolipin, glycoprotein 2, glycoprotein 210, heparin, heparan sulphate, histone H2A, prothrombin protein and vimentin) and 12 IgM (amyloid, cardiolipin, centromere protein A, collagen II, histones H2A and H2B, heparan sulphate, heparin, mitochondrial 2, nuclear Mi-2, nucleoporin 62 and vimentin) autoAbs were present at significantly different levels in NPSLE. The combination of IgG autoAbs against heparan sulphate, histone H2B and vimentin could differentiate NPSLE from non-NPSLE (area under the curve 0.845, 99.97% CI: 0.756, 0.933; P < 0.0001). Compared with non-NPSLE, four IgG and seven IgM autoAbs were significantly associated with inflammatory NPSLE. In ischaemic NPSLE, three IgG and three IgM autoAbs were significantly different from non-NPSLE patients. Conclusion In our cohort, the presence of high levels of anti-heparan sulphate and anti-histone H2B combined with low levels of anti-vimentin IgG autoAbs is highly suggestive of NPSLE. These results need to be validated in external cohorts.
Collapse
Affiliation(s)
- Pomme M van der Meulen
- Department of Pediatric Hematology, Immunology and Infectious Diseases, Emma Children's Hospital Academic Medical Center
| | - Anouk M Barendregt
- Department of Pediatric Hematology, Immunology and Infectious Diseases, Emma Children's Hospital Academic Medical Center
| | - Eloy Cuadrado
- Astrocyte Biology and Neurodegeneration Group, Netherlands Institute for Neuroscience, Amsterdam
| | - César Magro-Checa
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Gerda M Steup-Beekman
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Dieneke Schonenberg-Meinema
- Department of Pediatric Hematology, Immunology and Infectious Diseases, Emma Children's Hospital Academic Medical Center
| | - J Merlijn Van den Berg
- Department of Pediatric Hematology, Immunology and Infectious Diseases, Emma Children's Hospital Academic Medical Center
| | - Quan-Zhen Li
- Department of Immunology and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Paul A Baars
- Department of Experimental Immunology, Academic Medical Center, Amsterdam
| | | | | | - Ineke R J M Ten Berge
- Department of Internal Medicine, Clinical Immunology & Nephrology, Academic Medical Center, Amsterdam, The Netherlands
| | - Tom W J Huizinga
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Taco W Kuijpers
- Department of Pediatric Hematology, Immunology and Infectious Diseases, Emma Children's Hospital Academic Medical Center
| |
Collapse
|
118
|
Morris DC, Cheung WL, Loi R, Zhang T, Lu M, Zhang ZG, Chopp M. Thymosin β4 for the treatment of acute stroke in aged rats. Neurosci Lett 2017; 659:7-13. [PMID: 28864242 DOI: 10.1016/j.neulet.2017.08.064] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 08/10/2017] [Accepted: 08/28/2017] [Indexed: 12/21/2022]
Abstract
Thymosin β4 (Tβ4) is a 5K peptide which influences cellular migration by inhibiting organization of the actin-cytoskeleton. Tβ4 has neurorestorative properties and is a potential candidate for the treatment of sub-acute stroke. Previous research demonstrated that Tβ4 improved neurological outcome in a young (3 months) rat model of embolic stroke. We hypothesized that Tβ4 would improve neurological outcome in an aged rat model of embolic stroke when administered 24h after embolic stroke. Aged Male Wistar rats (Charles River, France 18-21 months) were subjected to embolic middle cerebral artery occlusion (MCAo). Rats were randomized to receive Tβ4 (12mg/kg, RegeneRx Biopharmaceuticals, Inc.) or control 24h after MCAo and then every 3days for 4 additional doses. The dose of 12mg/kg was the maximal dose of Tβ4 that showed functional improvement in a young rat model of embolic stroke. Functional tests (adhesive-removal test (ART), foot fault test (FFT) and the modified Neurological Severity Score (mNSS)) were performed weekly. The rats were sacrificed 56days after MCAo and lesion volumes were measured. Immunohistochemical analysis for oligodendrogenesis, myelination and gliosis was also performed. Twenty-three rats were included in the study: control group (n=12) and Tβ4 group (n=11). After randomization, there were three deaths in both the control and Tβ4 groups. The Tβ4 treatment reduced infarct volume by more than 50% (12.8%±9.3%, mean±SE, p<0.05) compared to the control group (26.0%±4.3%). However, Tβ4 did not show improvement in functional outcome compared to control. There was no significant increase in oligodendrogenesis, myelination and gliosis between control and treatment with Tβ4, however, we unexpectedly observed that overall (control and Tβ4 groups) astrocytic gliosis as measured by GFAP immunoreactivity was significantly inversely correlated with neurological outcome measured using the modified Neurological Severity Score (mNSS) (p<0.01), suggesting that greater gliosis may be related to improvement of neurological outcome in aged rats. In summary, Tβ4 treatment of stroke aged rats significantly reduces infarct volume compared to vehicle treated stroke, however, Tβ4 treatment did not show improvement in functional outcome, myelination or gliosis when compared to control. GFAP staining was significantly inversely correlated to improvement in the mNSS, suggesting that gliosis in the aged rat may be of benefit in improvement of functional outcome.
Collapse
Affiliation(s)
- Daniel C Morris
- Department of Emergency Medicine, Henry Ford Health System, Detroit, MI 48202, USA.
| | - Wing Lee Cheung
- Department of Emergency Medicine, Henry Ford Health System, Detroit, MI 48202, USA.
| | - Richard Loi
- Department of Emergency Medicine, Henry Ford Health System, Detroit, MI 48202, USA
| | - Talan Zhang
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI 48202, USA.
| | - Mei Lu
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI 48202, USA.
| | - Zheng G Zhang
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA.
| | - Michael Chopp
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; Department of Physics, Oakland University, Rochester, MI 48309, USA.
| |
Collapse
|
119
|
Sun L, Min L, Zhou H, Li M, Shao F, Wang W. Adolescent social isolation affects schizophrenia-like behavior and astrocyte biomarkers in the PFC of adult rats. Behav Brain Res 2017; 333:258-266. [DOI: 10.1016/j.bbr.2017.07.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Revised: 07/05/2017] [Accepted: 07/09/2017] [Indexed: 10/19/2022]
|
120
|
Laterza C, Wattananit S, Uoshima N, Ge R, Pekny R, Tornero D, Monni E, Lindvall O, Kokaia Z. Monocyte depletion early after stroke promotes neurogenesis from endogenous neural stem cells in adult brain. Exp Neurol 2017; 297:129-137. [PMID: 28746827 DOI: 10.1016/j.expneurol.2017.07.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 06/23/2017] [Accepted: 07/20/2017] [Indexed: 12/20/2022]
Abstract
Ischemic stroke, caused by middle cerebral artery occlusion, leads to long-lasting formation of new striatal neurons from neural stem/progenitor cells (NSPCs) in the subventricular zone (SVZ) of adult rodents. Concomitantly with this neurogenic response, SVZ exhibits activation of resident microglia and infiltrating monocytes. Here we show that depletion of circulating monocytes, using the anti-CCR2 antibody MC-21 during the first week after stroke, enhances striatal neurogenesis at one week post-insult, most likely by increasing short-term survival of the newly formed neuroblasts in the SVZ and adjacent striatum. Blocking monocyte recruitment did not alter the volume of the ischemic lesion but gave rise to reduced astrocyte activation in SVZ and adjacent striatum, which could contribute to the improved neuroblast survival. A similar decrease of astrocyte activation was found in and around human induced pluripotent stem cell (iPSC)-derived NSPCs transplanted into striatum at one week after stroke in monocyte-depleted mice. However, there was no effect on neurogenesis in the graft as determined 8weeks after implantation. Our findings demonstrate, for the first time, that a specific cellular component of the early inflammatory reaction in SVZ and adjacent striatum following stroke, i.e., infiltrating monocytes, compromises the short-term neurogenic response neurogenesis from endogenous NSPCs.
Collapse
Affiliation(s)
- Cecilia Laterza
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, University Hospital, SE-221 84 Lund, Sweden
| | - Somsak Wattananit
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, University Hospital, SE-221 84 Lund, Sweden
| | - Naomi Uoshima
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, University Hospital, SE-221 84 Lund, Sweden; Department of Anesthesiology, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Ruimin Ge
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, University Hospital, SE-221 84 Lund, Sweden
| | - Roy Pekny
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, University Hospital, SE-221 84 Lund, Sweden
| | - Daniel Tornero
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, University Hospital, SE-221 84 Lund, Sweden
| | - Emanuela Monni
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, University Hospital, SE-221 84 Lund, Sweden
| | - Olle Lindvall
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, University Hospital, SE-221 84 Lund, Sweden
| | - Zaal Kokaia
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, University Hospital, SE-221 84 Lund, Sweden.
| |
Collapse
|
121
|
Yoon H, Walters G, Paulsen AR, Scarisbrick IA. Astrocyte heterogeneity across the brain and spinal cord occurs developmentally, in adulthood and in response to demyelination. PLoS One 2017; 12:e0180697. [PMID: 28700615 PMCID: PMC5507262 DOI: 10.1371/journal.pone.0180697] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 06/20/2017] [Indexed: 01/08/2023] Open
Abstract
Astrocytes have emerged as essential regulators of function and response to injury in the brain and spinal cord, yet very little is known about regional differences that exist. Here we compare the expression of key astroglial markers (glial fibrillary acidic protein (GFAP) and Aldehyde Dehydrogenase-1 Family Member L1 (ALDH1L1)) across these disparate poles of the neuraxis, tracking their expression developmentally and in the context of demyelination. In addition, we document changes in the astrocyte regulatory cytokine interleukin 6 (IL-6), and its signaling partner signal transducer and activator of transcription 3 (STAT3), in vivo and in vitro. Results demonstrate that GFAP expression is higher in the developing and adult spinal cord relative to brain. Comparisons between GFAP and ALDH1L1 expression suggest elevations in spinal cord GFAP during the early postnatal period reflect an accelerated appearance of astrocytes, while elevations in adulthood reflect higher expression by individual astrocytes. Notably, increases in spinal cord compared to whole brain GFAP were paralleled by higher levels of IL-6 and STAT3. Equivalent elevations in GFAP, GFAP/ALDH1L1 ratios, and in IL-6, were observed in primary astrocyte cultures derived from spinal cord compared to cortex. Also, higher levels of GFAP were observed in the spinal cord compared to the brain after focal demyelinating injury. Altogether, these studies point to key differences in astrocyte abundance and the expression of GFAP and IL-6 across the brain and spinal cord that are positioned to influence regional specialization developmentally and responses occurring in the context of injury and disease.
Collapse
Affiliation(s)
- Hyesook Yoon
- Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Grant Walters
- Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Alex R. Paulsen
- Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Isobel A. Scarisbrick
- Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States of America
- Neurobiology of Disease Program, Mayo Clinic, Rochester, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
122
|
|
123
|
Reactive astrogliosis in stroke: Contributions of astrocytes to recovery of neurological function. Neurochem Int 2017; 107:88-103. [PMID: 28057555 DOI: 10.1016/j.neuint.2016.12.016] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 12/26/2016] [Accepted: 12/30/2016] [Indexed: 12/31/2022]
Abstract
Alterations in neuronal connectivity, particularly in the "peri-infarct" tissue adjacent to the region of ischemic damage, are important contributors to the spontaneous recovery of function that commonly follows stroke. Peri-infarct astrocytes undergo reactive astrogliosis and play key roles in modulating the adaptive responses in neurons. This reactive astrogliosis shares many features with that induced by other forms of damage to the central nervous system but also differs in details that potentially influence neurological recovery. A subpopulation of astrocytes within a few hundred micrometers of the infarct proliferate and are centrally involved in the development of the glial scar that separates the damaged tissue in the infarct from surrounding normal brain. The intertwined processes of astrocytes adjacent to the infarct provide the core structural component of the mature scar. Interventions that cause early disruption of glial scar formation typically impede restoration of neurological function. Marked reactive astrogliosis also develops in cells more distant from the infarct but these cells largely remain in the spatial territories they occupied prior to stroke. These cells play important roles in controlling the extracellular environment and release proteins and other molecules that are able to promote neuronal plasticity and improve functional recovery. Treatments manipulating aspects of reactive astrogliosis can enhance neuronal plasticity following stroke. Optimising these treatments for use in human stroke would benefit from a more complete characterization of the specific responses of peri-infarct astrocytes to stroke as well as a better understanding of the influence of other factors including age, sex, comorbidities and reperfusion of the ischemic tissue.
Collapse
|
124
|
|
125
|
Okabe N, Narita K, Miyamoto O. Axonal remodeling in the corticospinal tract after stroke: how does rehabilitative training modulate it? Neural Regen Res 2017; 12:185-192. [PMID: 28400791 PMCID: PMC5361493 DOI: 10.4103/1673-5374.200792] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Stroke causes long-term disability, and rehabilitative training is commonly used to improve the consecutive functional recovery. Following brain damage, surviving neurons undergo morphological alterations to reconstruct the remaining neural network. In the motor system, such neural network remodeling is observed as a motor map reorganization. Because of its significant correlation with functional recovery, motor map reorganization has been regarded as a key phenomenon for functional recovery after stroke. Although the mechanism underlying motor map reorganization remains unclear, increasing evidence has shown a critical role for axonal remodeling in the corticospinal tract. In this study, we review previous studies investigating axonal remodeling in the corticospinal tract after stroke and discuss which mechanisms may underlie the stimulatory effect of rehabilitative training. Axonal remodeling in the corticospinal tract can be classified into three types based on the location and the original targets of corticospinal neurons, and it seems that all the surviving corticospinal neurons in both ipsilesional and contralesional hemisphere can participate in axonal remodeling and motor map reorganization. Through axonal remodeling, corticospinal neurons alter their output selectivity from a single to multiple areas to compensate for the lost function. The remodeling of the corticospinal axon is influenced by the extent of tissue destruction and promoted by various therapeutic interventions, including rehabilitative training. Although the precise molecular mechanism underlying rehabilitation-promoted axonal remodeling remains elusive, previous data suggest that rehabilitative training promotes axonal remodeling by upregulating growth-promoting and downregulating growth-inhibiting signals.
Collapse
Affiliation(s)
- Naohiko Okabe
- Second Department of Physiology, Kawasaki Medical School 577, Matsushima, Kurashiki City, Okayama, Japan
| | - Kazuhiko Narita
- Second Department of Physiology, Kawasaki Medical School 577, Matsushima, Kurashiki City, Okayama, Japan
| | - Osamu Miyamoto
- Second Department of Physiology, Kawasaki Medical School 577, Matsushima, Kurashiki City, Okayama, Japan
| |
Collapse
|
126
|
Magistri M, Khoury N, Mazza EMC, Velmeshev D, Lee JK, Bicciato S, Tsoulfas P, Faghihi MA. A comparative transcriptomic analysis of astrocytes differentiation from human neural progenitor cells. Eur J Neurosci 2016; 44:2858-2870. [PMID: 27564458 DOI: 10.1111/ejn.13382] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 07/25/2016] [Accepted: 08/23/2016] [Indexed: 12/11/2022]
Abstract
Astrocytes are a morphologically and functionally heterogeneous population of cells that play critical roles in neurodevelopment and in the regulation of central nervous system homeostasis. Studies of human astrocytes have been hampered by the lack of specific molecular markers and by the difficulties associated with purifying and culturing astrocytes from adult human brains. Human neural progenitor cells (NPCs) with self-renewal and multipotent properties represent an appealing model system to gain insight into the developmental genetics and function of human astrocytes, but a comprehensive molecular characterization that confirms the validity of this cellular system is still missing. Here we used an unbiased transcriptomic analysis to characterize in vitro culture of human NPCs and to define the gene expression programs activated during the differentiation of these cells into astrocytes using FBS or the combination of CNTF and BMP4. Our results demonstrate that in vitro cultures of human NPCs isolated during the gliogenic phase of neurodevelopment mainly consist of radial glial cells (RGCs) and glia-restricted progenitor cells. In these cells the combination of CNTF and BMP4 activates the JAK/STAT and SMAD signaling cascades, leading to the inhibition of oligodendrocytes lineage commitment and activation of astrocytes differentiation. On the other hand, FBS-derived astrocytes have properties of reactive astrocytes. Our work suggests that in vitro culture of human NPCs represents a valuable cellular system to study human disorders characterized by impairment of astrocytes development and function. Our datasets represent an important resource for researchers studying human astrocytes development and might set the basis for the discovery of novel human-specific astrocyte markers.
Collapse
Affiliation(s)
- Marco Magistri
- Department of Psychiatry and Behavioral Sciences, Center for Therapeutic Innovation, University of Miami Miller School of Medicine, 1501 NW 10th Ave, BRB 508, Miami, FL, 33136, USA
| | - Nathalie Khoury
- Department of Psychiatry and Behavioral Sciences, Center for Therapeutic Innovation, University of Miami Miller School of Medicine, 1501 NW 10th Ave, BRB 508, Miami, FL, 33136, USA
| | - Emilia Maria Cristina Mazza
- Department of Life Sciences, Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy
| | - Dmitry Velmeshev
- Department of Psychiatry and Behavioral Sciences, Center for Therapeutic Innovation, University of Miami Miller School of Medicine, 1501 NW 10th Ave, BRB 508, Miami, FL, 33136, USA
| | - Jae K Lee
- Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Silvio Bicciato
- Department of Life Sciences, Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy
| | - Pantelis Tsoulfas
- Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Mohammad Ali Faghihi
- Department of Psychiatry and Behavioral Sciences, Center for Therapeutic Innovation, University of Miami Miller School of Medicine, 1501 NW 10th Ave, BRB 508, Miami, FL, 33136, USA
| |
Collapse
|
127
|
Radulovic M, Yoon H, Wu J, Mustafa K, Scarisbrick IA. Targeting the thrombin receptor modulates inflammation and astrogliosis to improve recovery after spinal cord injury. Neurobiol Dis 2016; 93:226-42. [PMID: 27145117 PMCID: PMC4930708 DOI: 10.1016/j.nbd.2016.04.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 04/08/2016] [Accepted: 04/29/2016] [Indexed: 02/07/2023] Open
Abstract
The deregulation of serine protease activity is a common feature of neurological injury, but little is known regarding their mechanisms of action or whether they can be targeted to facilitate repair. In this study we demonstrate that the thrombin receptor (Protease Activated Receptor 1, (PAR1)) serves as a critical translator of the spinal cord injury (SCI) proteolytic microenvironment into a cascade of pro-inflammatory events that contribute to astrogliosis and functional decline. PAR1 knockout mice displayed improved locomotor recovery after SCI and reduced signatures of inflammation and astrogliosis, including expression of glial fibrillary acidic protein (GFAP), vimentin, and STAT3 signaling. SCI-associated elevations in pro-inflammatory cytokines such as IL-1β and IL-6 were also reduced in PAR1-/- mice and co-ordinate improvements in tissue sparing and preservation of NeuN-positive ventral horn neurons, and PKCγ corticospinal axons, were observed. PAR1 and its agonist's thrombin and neurosin were expressed by perilesional astrocytes and each agonist increased the production of IL-6 and STAT3 signaling in primary astrocyte cultures in a PAR1-dependent manner. In turn, IL-6-stimulated astrocytes increased expression of PAR1, thrombin, and neurosin, pointing to a model in which PAR1 activation contributes to increased astrogliosis by feedforward- and feedback-signaling dynamics. Collectively, these findings identify the thrombin receptor as a key mediator of inflammation and astrogliosis in the aftermath of SCI that can be targeted to reduce neurodegeneration and improve neurobehavioral recovery.
Collapse
Affiliation(s)
- Maja Radulovic
- Neurobiology of Disease Program, Mayo Medical and Graduate School, Rehabilitation Medicine Research Center, Rochester 55905, MN, United States
| | - Hyesook Yoon
- Department of Physical Medicine and Rehabilitation, Mayo Medical and Graduate School, Rehabilitation Medicine Research Center, Rochester, MN 55905, United States; Department of Physiology and Biomedical Engineering, Mayo Medical and Graduate School, Rehabilitation Medicine Research Center, Rochester, MN 55905, United States
| | - Jianmin Wu
- Department of Physical Medicine and Rehabilitation, Mayo Medical and Graduate School, Rehabilitation Medicine Research Center, Rochester, MN 55905, United States
| | - Karim Mustafa
- Neurobiology of Disease Program, Mayo Medical and Graduate School, Rehabilitation Medicine Research Center, Rochester 55905, MN, United States
| | - Isobel A Scarisbrick
- Neurobiology of Disease Program, Mayo Medical and Graduate School, Rehabilitation Medicine Research Center, Rochester 55905, MN, United States; Department of Physical Medicine and Rehabilitation, Mayo Medical and Graduate School, Rehabilitation Medicine Research Center, Rochester, MN 55905, United States; Department of Physiology and Biomedical Engineering, Mayo Medical and Graduate School, Rehabilitation Medicine Research Center, Rochester, MN 55905, United States.
| |
Collapse
|
128
|
Egawa N, Lok J, Washida K, Arai K. Mechanisms of Axonal Damage and Repair after Central Nervous System Injury. Transl Stroke Res 2016; 8:14-21. [PMID: 27566737 DOI: 10.1007/s12975-016-0495-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 08/14/2016] [Accepted: 08/16/2016] [Indexed: 01/19/2023]
Abstract
Central nervous system (CNS) injury initiates spatial and temporal neurodegeneration. Under pathologic conditions, damaged glial cells cannot supply sufficient metabolites to neurons, leading to energy deficiency for neuronal axons. The widespread disruption of cellular membranes causes disturbed intracellular signaling via dysregulated ionic gradients in neurons. Although several deleterious cascades are activated during the acute phase of CNS injury, some compensatory responses may tend to promote axonal repair during the chronic/remodeling phase. Because it may not be easy to block all multifactorial neurodegenerative pathways after CNS injury, supporting or boosting endogenous regenerative mechanisms would be an important therapeutic approach for CNS diseases. In this mini-review, we briefly but broadly introduce basic mechanisms that trigger axonal degeneration and then discuss potential targets for promoting axonal regeneration after CNS injury.
Collapse
Affiliation(s)
- Naohiro Egawa
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, MGH East 149-2401, Charlestown, MA, 02129, USA
| | - Josephine Lok
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, MGH East 149-2401, Charlestown, MA, 02129, USA.,Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Kazuo Washida
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, MGH East 149-2401, Charlestown, MA, 02129, USA
| | - Ken Arai
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, MGH East 149-2401, Charlestown, MA, 02129, USA.
| |
Collapse
|
129
|
Hao XZ, Yin LK, Zhang XX, Tian JQ, Li CC, Feng XY, Jiang M, Yang YM. Combining systemic and stereotactic MEMRI to detect the correlation between gliosis and neuronal connective pathway at the chronic stage after stroke. J Neuroinflammation 2016; 13:156. [PMID: 27316350 PMCID: PMC4912752 DOI: 10.1186/s12974-016-0622-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 06/10/2016] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND The early dysfunction and subsequent recovery after stroke, characterized by the destruction and remodeling of connective pathways between cortex and subcortical regions, is associated with neuroinflammation. As major components of the inflammatory process, reactive astrocytes have double-edged effects on pathological progression. The temporal patterns of astrocyte and neuronal pathway activity can be revealed by systemic and stereotactic manganese-enhanced magnetic resonance imaging (MEMRI), respectively. In the present study, we aimed to detect an association between astrocyte activity and recovery of neuronal connective pathways by combining systemic with stereotactic MEMRI. METHODS Fifty adult rats, divided into two groups, underwent a 60-min occlusion of the middle cerebral artery. The groups were given either a systemic administration or stereotactic injection of MnCl2 at 1, 3, 7, and 14 days after stroke and underwent MRI 4 and 2 days later, respectively. Immunofluorescence (IF) of group 1 was conducted to corroborate the results. Repetitive behavioral testing was also performed with all rats at 1, 3, 7, and 14 days to obtain a functional score. RESULTS Ring- or crescent-shaped enhancements formed in the striatal peri-infarct regions (STR) at 11 and 18 days. This was concurrent with the activity of glial fibrillary acidic protein (GFAP)-positive astrocytes, which mainly localized at the peri-infarct region and significantly increased in number at 11 and 18 days after stroke. Microglia/macrophages, detected by IF, mainly localized in the lesion core, rather than in the region of enhancement. The ipsilateral substantia nigra (SN) revealed Mn-related signal enhancement reduction and subsequent signs of the recovery process at 3 to 5 days and 9 to 16 days, respectively. Behavioral testing showed that sensorimotor functions were initially disturbed, but subsequently recovered at 7 and 14 days. CONCLUSIONS We found a positive temporal correlation between astrogliosis and the recovery of neuronal connective pathways at the chronic stage by using the in vivo method of MEMRI. Our results highlighted the potential contribution of astrocytes to the neuronal recovery of these connective pathways.
Collapse
Affiliation(s)
- Xiao-zhu Hao
- />Department of Radiology, Huashan Hospital, Fudan University, Shanghai, 200040 China
| | - Le-kang Yin
- />Department of Radiology, Huashan Hospital, Fudan University, Shanghai, 200040 China
| | - Xiao-xue Zhang
- />Department of Radiology, Huashan Hospital, Fudan University, Shanghai, 200040 China
| | - Jia-qi Tian
- />Department of Radiology, Huashan Hospital, Fudan University, Shanghai, 200040 China
| | - Chan-chan Li
- />Department of Radiology, Huashan Hospital, Fudan University, Shanghai, 200040 China
| | - Xiao-yuan Feng
- />Department of Radiology, Huashan Hospital, Fudan University, Shanghai, 200040 China
| | - Min Jiang
- />Institutes of Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200032 China
| | - Yan-mei Yang
- />Department of Radiology, Huashan Hospital, Fudan University, Shanghai, 200040 China
| |
Collapse
|
130
|
Abstract
Astrocytes are the most explored non-neuronal cells in the brain under neurophysiological and neurodegenerative conditions. Extensive research has been done to understand their specific role during neuropathological conditions but still the existing findings could not conclude their mechanism of action and their specific role in neurodegenerative conditions. This review discusses their physiological and pathological roles, their activation, morphological alterations and their probable use in search of new therapeutic targets for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Sarika Singh
- a 1 Toxicology Division, CSIR-CDRI , Lucknow , India.,b 2 Department of Biochemistry and Biophysics , University of California , San Francisco, San Francisco , CA , USA
| | - Neeraj Joshi
- a 1 Toxicology Division, CSIR-CDRI , Lucknow , India.,b 2 Department of Biochemistry and Biophysics , University of California , San Francisco, San Francisco , CA , USA
| |
Collapse
|
131
|
Dang G, Chen X, Chen Y, Zhao Y, Ouyang F, Zeng J. Dynamic secondary degeneration in the spinal cord and ventral root after a focal cerebral infarction among hypertensive rats. Sci Rep 2016; 6:22655. [PMID: 26949108 PMCID: PMC4780069 DOI: 10.1038/srep22655] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 02/18/2016] [Indexed: 01/22/2023] Open
Abstract
Cerebral infarction can cause secondary damage to nonischemic brain regions. However, whether this phenomenon will appear in central nervous system regions outside the brain remains unclear. Here we investigated pathological changes in the spinal cord and ventral root after ischemic stroke. All rats exhibited apparent neurological deficits post-MCAO, which improved gradually but could still be detected 12-weeks. Neuronal filaments in the corticospinal tract (CST) and neurons in the ventral horn were significantly declined in the contralateral cervical and lumbar enlargement 1-week post-MCAO. These decreases remained stable until 12-weeks, accompanied by progressively increased glial activation in the ventral horn. Axonal degeneration and structural derangement were evident in the contralateral cervical and lumbar ventral root 1-week post-MCAO; these changes spontaneously attenuated over time, but abnormalities could still be observed 12-weeks. The number of neural fibers in the contralateral CST and neurons in the contralateral ventral horn were positively correlated with neurological scores 12-weeks post-MCAO. Additionally, GFAP+cell density in the contralateral CST and ventral horn was negatively correlated with neurological scores. Our results suggest that cerebral infarction can elicit secondary degeneration in the cervical and lumbar spinal cord, as well as the projecting ventral root, which may hamper functional recovery after stroke.
Collapse
Affiliation(s)
- Ge Dang
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Xinran Chen
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Yicong Chen
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Yuhui Zhao
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Fubing Ouyang
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Jinsheng Zeng
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| |
Collapse
|
132
|
Plasticity in the Neonatal Brain following Hypoxic-Ischaemic Injury. Neural Plast 2016; 2016:4901014. [PMID: 27047695 PMCID: PMC4800097 DOI: 10.1155/2016/4901014] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 01/12/2016] [Accepted: 02/07/2016] [Indexed: 12/03/2022] Open
Abstract
Hypoxic-ischaemic damage to the developing brain is a leading cause of child death, with high mortality and morbidity, including cerebral palsy, epilepsy, and cognitive disabilities. The developmental stage of the brain and the severity of the insult influence the selective regional vulnerability and the subsequent clinical manifestations. The increased susceptibility to hypoxia-ischaemia (HI) of periventricular white matter in preterm infants predisposes the immature brain to motor, cognitive, and sensory deficits, with cognitive impairment associated with earlier gestational age. In term infants HI causes selective damage to sensorimotor cortex, basal ganglia, thalamus, and brain stem. Even though the immature brain is more malleable to external stimuli compared to the adult one, a hypoxic-ischaemic event to the neonate interrupts the shaping of central motor pathways and can affect normal developmental plasticity through altering neurotransmission, changes in cellular signalling, neural connectivity and function, wrong targeted innervation, and interruption of developmental apoptosis. Models of neonatal HI demonstrate three morphologically different types of cell death, that is, apoptosis, necrosis, and autophagy, which crosstalk and can exist as a continuum in the same cell. In the present review we discuss the mechanisms of HI injury to the immature brain and the way they affect plasticity.
Collapse
|
133
|
Reactive gliosis in the pathogenesis of CNS diseases. Biochim Biophys Acta Mol Basis Dis 2016; 1862:483-91. [DOI: 10.1016/j.bbadis.2015.11.014] [Citation(s) in RCA: 157] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 11/19/2015] [Accepted: 11/30/2015] [Indexed: 01/11/2023]
|
134
|
Abeysinghe HCS, Phillips EL, Chin-Cheng H, Beart PM, Roulston CL. Modulating Astrocyte Transition after Stroke to Promote Brain Rescue and Functional Recovery: Emerging Targets Include Rho Kinase. Int J Mol Sci 2016; 17:288. [PMID: 26927079 PMCID: PMC4813152 DOI: 10.3390/ijms17030288] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 01/26/2016] [Accepted: 02/05/2016] [Indexed: 01/13/2023] Open
Abstract
Stroke is a common and serious condition, with few therapies. Whilst previous focus has been directed towards biochemical events within neurons, none have successfully prevented the progression of injury that occurs in the acute phase. New targeted treatments that promote recovery after stroke might be a better strategy and are desperately needed for the majority of stroke survivors. Cells comprising the neurovascular unit, including blood vessels and astrocytes, present an alternative target for supporting brain rescue and recovery in the late phase of stroke, since alteration in the unit also occurs in regions outside of the lesion. One of the major changes in the unit involves extensive morphological transition of astrocytes resulting in altered energy metabolism, decreased glutamate reuptake and recycling, and retraction of astrocyte end feed from both blood vessels and neurons. Whilst globally inhibiting transitional change in astrocytes after stroke is reported to result in further damage and functional loss, we discuss the available evidence to suggest that the transitional activation of astrocytes after stroke can be modulated for improved outcomes. In particular, we review the role of Rho-kinase (ROCK) in reactive gliosis and show that inhibiting ROCK after stroke results in reduced scar formation and improved functional recovery.
Collapse
Affiliation(s)
- Hima Charika S Abeysinghe
- Neurotrauma Research, Department of Medicine, St Vincent's Campus, University of Melbourne, Parkville, VIC 3065, Australia.
- Department of Surgery, St Vincent's Campus, University of Melbourne, Parkville, VIC 3065, Australia.
| | - Ellie L Phillips
- Department of Biochemistry and Molecular Biology, Bio21 Insitute, University of Melbourne, Parkville, VIC 3010, Australia.
| | - Heung Chin-Cheng
- Department of Biochemistry and Molecular Biology, Bio21 Insitute, University of Melbourne, Parkville, VIC 3010, Australia.
| | - Philip M Beart
- The Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, Parkville, VIC 3010, Australia.
| | - Carli L Roulston
- Neurotrauma Research, Department of Medicine, St Vincent's Campus, University of Melbourne, Parkville, VIC 3065, Australia.
| |
Collapse
|
135
|
Zhang C, Meng Q, Zhang X, Wu S, Wang S, Chen R, Li X. Role of astrocyte activation in fine particulate matter-enhancement of existing ischemic stroke in Sprague-Dawley male rats. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2016; 79:393-401. [PMID: 27267821 DOI: 10.1080/15287394.2016.1176615] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Exposure to particulate matter (PM) with an aerodynamic diameter of less than 2.5 μm (PM2.5) is associated with increased risk of ischemic stroke, but potential neurotoxic mechanisms remain to be determined. In this study, adult male Sprague- Dawley (SD) rats were divided into four groups as follows: control (CON), PM2.5 exposure (PM alone), ischemic stroke (IS), and ischemic stroke and PM2.5 (IS-PM). Ischemic stroke groups were prepared by middle cerebral artery occlusion (MCAO), and neurobehavior was assessed daily for 7 consecutive days. The control group was administered intranasally 20 μl PBS, while PM2.5 alone was given as 20 μl of PM2.5 (10 mg/ml) intranasal daily for 7 consecutive days. The spontaneous locomotion and exploratory behavior of rats were assessed by the open field test. Cells positive for glial fibrillary acidic protein (GFAP) and inducible nitric oxide synthase (iNOS) were determined for astrocyte activation and inflammatory reactions. Neuronal edema and pyknosis in the cerebral cortex, hippocampus, and midbrain were observed in IS groups with or without PM2.5 treatment. Astrocyte activity was enhanced, whereas spontaneous locomotion and exploratory movements decreased in the IS-PM group. Data demonstrated that astrocytes activation and inflammatory reactions may play a role in IS and that exposure to PM2.5 may aggravate the neurobehavioral alterations observed in rats suffering from IS.
Collapse
Affiliation(s)
- Chengcheng Zhang
- a Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health , Southeast University , Nanjing 210009 , China
| | - Qingtao Meng
- a Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health , Southeast University , Nanjing 210009 , China
| | - Xin Zhang
- a Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health , Southeast University , Nanjing 210009 , China
| | - Shenshen Wu
- a Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health , Southeast University , Nanjing 210009 , China
| | - Shizhi Wang
- a Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health , Southeast University , Nanjing 210009 , China
| | - Rui Chen
- a Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health , Southeast University , Nanjing 210009 , China
| | - Xiaobo Li
- a Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health , Southeast University , Nanjing 210009 , China
| |
Collapse
|
136
|
Chisholm NC, Sohrabji F. Astrocytic response to cerebral ischemia is influenced by sex differences and impaired by aging. Neurobiol Dis 2016; 85:245-253. [PMID: 25843666 PMCID: PMC5636213 DOI: 10.1016/j.nbd.2015.03.028] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Revised: 03/16/2015] [Accepted: 03/26/2015] [Indexed: 12/21/2022] Open
Abstract
Ischemic stroke occurs more often among the elderly, and within this demographic, women are at an increased risk for stroke and have poorer functional recovery than men. This is also well replicated in animal studies where aging females are shown to have more extensive brain tissue loss as compared to adult females. Astrocytes provide nutrients for neurons, regulate glutamate levels, and release neurotrophins and thus play a key role in the events that occur following ischemia. In addition, astrocytes express receptors for gonadal hormones and synthesize several neurosteroids suggesting that the sex differences in stroke outcome may be mediated through astrocytes. This review discusses key astrocytic responses to ischemia including, reactive gliosis, excitotoxicity, and neuroinflammation. In light of the age and sex differences in stroke outcomes, this review highlights how aging and gonadal hormones influence these responses. Lastly, astrocyte specific changes in gene expression and epigenetic modifications during aging and following ischemia are discussed as possible molecular mechanisms for impaired astrocytic functioning.
Collapse
Affiliation(s)
- Nioka C Chisholm
- Department of Neuroscience and Experimental Therapeutics, Texas A & M Health Science Center, College of Medicine, Bryan, TX 77807, USA
| | - Farida Sohrabji
- Department of Neuroscience and Experimental Therapeutics, Texas A & M Health Science Center, College of Medicine, Bryan, TX 77807, USA.
| |
Collapse
|
137
|
Electro-acupuncture at LI11 and ST36 acupoints exerts neuroprotective effects via reactive astrocyte proliferation after ischemia and reperfusion injury in rats. Brain Res Bull 2015; 120:14-24. [PMID: 26524137 DOI: 10.1016/j.brainresbull.2015.10.011] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Revised: 10/22/2015] [Accepted: 10/26/2015] [Indexed: 11/23/2022]
Abstract
Reactive astrogliosis is a common phenomenon in central nervous system (CNS) injuries such as ischemic stroke. The present study aimed to deeply investigate the relationships between the neuroprotective effect of electro-acupuncture (EA) and reactive astrocytes following cerebral ischemia. EA treatment at the Quchi (LI11) and Zusanli (ST36) acupoints at Day 3 attenuated neurological deficits and cerebral infarct volume in ischemia and reperfusion (I/R) injured rats. Animal behavior assessments found that the speed of Catwalk gait, equilibrium and coordination of Rotarod test were improved. Furthermore, EA treatment exerted neuroprotective effects via activation of glial fibrillary acidic protein (GFAP), vimentin and nestin positive cells. Simultaneously, an obvious increase in GFAP/vimentin, GFAP/nestin and GFAP/BrdU co-labeling appeared in the peri-infract cortex and striatum, suggesting EA can promote the proliferation of GFAP/vimentin/nestin-positive reactive astrocytes. The expression of cell cycle-associated proteins Cyclin Dl, CDK4 and phospho-Rb were increased in the peri-infract cortex and striatum, indicating proliferated reactive astrocytes-mediated CyclinDl/CDK4 regulation of the transition of the G1-to-S cell cycle phases. In addition, EA enhanced the localized expression of brain-derived neurotrophic factor (BDNF) in the peri-infract cortex and striatum. These results demonstrated that EA treatment at the LI11 and ST36 acupoints on Day 3 exerted neuroprotection via proliferation of GFAP/vimentin/nestin-positive reactive astrocytes and, potentially, secretion of reactive astrocytes-derived BDNF in I/R injured rats.
Collapse
|
138
|
Caleo M. Rehabilitation and plasticity following stroke: Insights from rodent models. Neuroscience 2015; 311:180-94. [PMID: 26493858 DOI: 10.1016/j.neuroscience.2015.10.029] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 10/11/2015] [Accepted: 10/12/2015] [Indexed: 01/08/2023]
Abstract
Ischemic injuries within the motor cortex result in functional deficits that may profoundly impact activities of daily living in patients. Current rehabilitation protocols achieve only limited recovery of motor abilities. The brain reorganizes spontaneously after injury, and it is believed that appropriately boosting these neuroplastic processes may restore function via recruitment of spared areas and pathways. Here I review studies on circuit reorganization, neuronal and glial plasticity and axonal sprouting following ischemic damage to the forelimb motor cortex, with a particular focus on rodent models. I discuss evidence pointing to compensatory take-over of lost functions by adjacent peri-lesional areas and the role of the contralesional hemisphere in recovery. One key issue is the need to distinguish "true" recovery (i.e. re-establishment of original movement patterns) from compensation in the assessment of post-stroke functional gains. I also consider the effects of physical rehabilitation, including robot-assisted therapy, and the potential mechanisms by which motor training induces recovery. Finally, I describe experimental approaches in which training is coupled with delivery of plasticizing drugs that render the remaining, undamaged pathways more sensitive to experience-dependent modifications. These combinatorial strategies hold promise for the definition of more effective rehabilitation paradigms that can be translated into clinical practice.
Collapse
Affiliation(s)
- M Caleo
- CNR Neuroscience Institute, via G. Moruzzi 1, 56124 Pisa, Italy; The BioRobotics Institute, Scuola Superiore Sant'Anna, P.zza Martiri della Libertà 33, 56127 Pisa, Italy.
| |
Collapse
|
139
|
Ergul A, Valenzuela JP, Fouda AY, Fagan SC. Cellular connections, microenvironment and brain angiogenesis in diabetes: Lost communication signals in the post-stroke period. Brain Res 2015; 1623:81-96. [PMID: 25749094 PMCID: PMC4743654 DOI: 10.1016/j.brainres.2015.02.045] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 02/18/2015] [Accepted: 02/23/2015] [Indexed: 12/16/2022]
Abstract
Diabetes not only increases the risk but also worsens the motor and cognitive recovery after stroke, which is the leading cause of disability worldwide. Repair after stroke requires coordinated communication among various cell types in the central nervous system as well as circulating cells. Vascular restoration is critical for the enhancement of neurogenesis and neuroplasticity. Given that vascular disease is a major component of all complications associated with diabetes including stroke, this review will focus on cellular communications that are important for vascular restoration in the context of diabetes. This article is part of a Special Issue entitled SI: Cell Interactions In Stroke.
Collapse
Affiliation(s)
- Adviye Ergul
- Charlie Norwood Veterans Affairs Medical Center, Augusta, GA 30904, USA; Department of Physiology, Medical College of Georgia, Georgia Regents University, 1120 15th Street, CA 2094, Augusta, GA 30912, USA; Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA.
| | - John Paul Valenzuela
- Department of Physiology, Medical College of Georgia, Georgia Regents University, 1120 15th Street, CA 2094, Augusta, GA 30912, USA
| | - Abdelrahman Y Fouda
- Charlie Norwood Veterans Affairs Medical Center, Augusta, GA 30904, USA; Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA
| | - Susan C Fagan
- Charlie Norwood Veterans Affairs Medical Center, Augusta, GA 30904, USA; Department of Neurology, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA; Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA
| |
Collapse
|
140
|
Liu Z, Chopp M. Astrocytes, therapeutic targets for neuroprotection and neurorestoration in ischemic stroke. Prog Neurobiol 2015; 144:103-20. [PMID: 26455456 DOI: 10.1016/j.pneurobio.2015.09.008] [Citation(s) in RCA: 412] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 08/06/2015] [Accepted: 09/05/2015] [Indexed: 01/04/2023]
Abstract
Astrocytes are the most abundant cell type within the central nervous system. They play essential roles in maintaining normal brain function, as they are a critical structural and functional part of the tripartite synapses and the neurovascular unit, and communicate with neurons, oligodendrocytes and endothelial cells. After an ischemic stroke, astrocytes perform multiple functions both detrimental and beneficial, for neuronal survival during the acute phase. Aspects of the astrocytic inflammatory response to stroke may aggravate the ischemic lesion, but astrocytes also provide benefit for neuroprotection, by limiting lesion extension via anti-excitotoxicity effects and releasing neurotrophins. Similarly, during the late recovery phase after stroke, the glial scar may obstruct axonal regeneration and subsequently reduce the functional outcome; however, astrocytes also contribute to angiogenesis, neurogenesis, synaptogenesis, and axonal remodeling, and thereby promote neurological recovery. Thus, the pivotal involvement of astrocytes in normal brain function and responses to an ischemic lesion designates them as excellent therapeutic targets to improve functional outcome following stroke. In this review, we will focus on functions of astrocytes and astrocyte-mediated events during stroke and recovery. We will provide an overview of approaches on how to reduce the detrimental effects and amplify the beneficial effects of astrocytes on neuroprotection and on neurorestoration post stroke, which may lead to novel and clinically relevant therapies for stroke.
Collapse
Affiliation(s)
- Zhongwu Liu
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA.
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA; Department of Physics, Oakland University, Rochester, MI, USA
| |
Collapse
|
141
|
Lebkuechner I, Wilhelmsson U, Möllerström E, Pekna M, Pekny M. Heterogeneity of Notch signaling in astrocytes and the effects of GFAP and vimentin deficiency. J Neurochem 2015; 135:234-48. [DOI: 10.1111/jnc.13213] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 06/09/2015] [Accepted: 06/11/2015] [Indexed: 11/29/2022]
Affiliation(s)
- Isabell Lebkuechner
- Center for Brain Repair and Rehabilitation; Department of Clinical Neuroscience and Rehabilitation; Institute of Neuroscience and Physiology; Sahlgrenska Academy at the University of Gothenburg; Gothenburg Sweden
| | - Ulrika Wilhelmsson
- Center for Brain Repair and Rehabilitation; Department of Clinical Neuroscience and Rehabilitation; Institute of Neuroscience and Physiology; Sahlgrenska Academy at the University of Gothenburg; Gothenburg Sweden
| | - Elin Möllerström
- Center for Brain Repair and Rehabilitation; Department of Clinical Neuroscience and Rehabilitation; Institute of Neuroscience and Physiology; Sahlgrenska Academy at the University of Gothenburg; Gothenburg Sweden
| | - Marcela Pekna
- Center for Brain Repair and Rehabilitation; Department of Clinical Neuroscience and Rehabilitation; Institute of Neuroscience and Physiology; Sahlgrenska Academy at the University of Gothenburg; Gothenburg Sweden
- Florey Institute of Neuroscience and Mental Health; Parkville Victoria Australia
- University of Newcastle; New South Wales Australia
| | - Milos Pekny
- Center for Brain Repair and Rehabilitation; Department of Clinical Neuroscience and Rehabilitation; Institute of Neuroscience and Physiology; Sahlgrenska Academy at the University of Gothenburg; Gothenburg Sweden
- Florey Institute of Neuroscience and Mental Health; Parkville Victoria Australia
- University of Newcastle; New South Wales Australia
| |
Collapse
|
142
|
Wunderlich KA, Tanimoto N, Grosche A, Zrenner E, Pekny M, Reichenbach A, Seeliger MW, Pannicke T, Perez MT. Retinal functional alterations in mice lacking intermediate filament proteins glial fibrillary acidic protein and vimentin. FASEB J 2015; 29:4815-28. [PMID: 26251181 DOI: 10.1096/fj.15-272963] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 07/27/2015] [Indexed: 01/02/2023]
Abstract
Vimentin (Vim) and glial fibrillary acidic protein (GFAP) are important components of the intermediate filament (IF) (or nanofilament) system of astroglial cells. We conducted full-field electroretinogram (ERG) recordings and found that whereas photoreceptor responses (a-wave) were normal in uninjured GFAP(-/-)Vim(-/-) mice, b-wave amplitudes were increased. Moreover, we found that Kir (inward rectifier K(+)) channel protein expression was reduced in the retinas of GFAP(-/-)Vim(-/-) mice and that Kir-mediated current amplitudes were lower in Müller glial cells isolated from these mice. Studies have shown that the IF system, in addition, is involved in the retinal response to injury and that attenuated Müller cell reactivity and reduced photoreceptor cell loss are observed in IF-deficient mice after experimental retinal detachment. We investigated whether the lack of IF proteins would affect cell survival in a retinal ischemia-reperfusion model. We found that although cell loss was induced in both genotypes, the number of surviving cells in the inner retina was lower in IF-deficient mice. Our findings thus show that the inability to produce GFAP and Vim affects normal retinal physiology and that the effect of IF deficiency on retinal cell survival differs, depending on the underlying pathologic condition.
Collapse
Affiliation(s)
- Kirsten A Wunderlich
- *Department of Clinical Sciences, Division of Ophthalmology, and NanoLund, Nanometer Structure Consortium, Lund University, Lund, Sweden; Graduate School of Cellular and Molecular Neuroscience, Center for Integrative Neuroscience (CIN), and Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany; Institute of Human Genetics, University of Regensburg, Regensburg, Germany; Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; **Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia; and Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany
| | - Naoyuki Tanimoto
- *Department of Clinical Sciences, Division of Ophthalmology, and NanoLund, Nanometer Structure Consortium, Lund University, Lund, Sweden; Graduate School of Cellular and Molecular Neuroscience, Center for Integrative Neuroscience (CIN), and Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany; Institute of Human Genetics, University of Regensburg, Regensburg, Germany; Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; **Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia; and Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany
| | - Antje Grosche
- *Department of Clinical Sciences, Division of Ophthalmology, and NanoLund, Nanometer Structure Consortium, Lund University, Lund, Sweden; Graduate School of Cellular and Molecular Neuroscience, Center for Integrative Neuroscience (CIN), and Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany; Institute of Human Genetics, University of Regensburg, Regensburg, Germany; Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; **Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia; and Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany
| | - Eberhart Zrenner
- *Department of Clinical Sciences, Division of Ophthalmology, and NanoLund, Nanometer Structure Consortium, Lund University, Lund, Sweden; Graduate School of Cellular and Molecular Neuroscience, Center for Integrative Neuroscience (CIN), and Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany; Institute of Human Genetics, University of Regensburg, Regensburg, Germany; Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; **Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia; and Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany
| | - Milos Pekny
- *Department of Clinical Sciences, Division of Ophthalmology, and NanoLund, Nanometer Structure Consortium, Lund University, Lund, Sweden; Graduate School of Cellular and Molecular Neuroscience, Center for Integrative Neuroscience (CIN), and Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany; Institute of Human Genetics, University of Regensburg, Regensburg, Germany; Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; **Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia; and Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany
| | - Andreas Reichenbach
- *Department of Clinical Sciences, Division of Ophthalmology, and NanoLund, Nanometer Structure Consortium, Lund University, Lund, Sweden; Graduate School of Cellular and Molecular Neuroscience, Center for Integrative Neuroscience (CIN), and Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany; Institute of Human Genetics, University of Regensburg, Regensburg, Germany; Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; **Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia; and Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany
| | - Mathias W Seeliger
- *Department of Clinical Sciences, Division of Ophthalmology, and NanoLund, Nanometer Structure Consortium, Lund University, Lund, Sweden; Graduate School of Cellular and Molecular Neuroscience, Center for Integrative Neuroscience (CIN), and Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany; Institute of Human Genetics, University of Regensburg, Regensburg, Germany; Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; **Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia; and Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany
| | - Thomas Pannicke
- *Department of Clinical Sciences, Division of Ophthalmology, and NanoLund, Nanometer Structure Consortium, Lund University, Lund, Sweden; Graduate School of Cellular and Molecular Neuroscience, Center for Integrative Neuroscience (CIN), and Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany; Institute of Human Genetics, University of Regensburg, Regensburg, Germany; Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; **Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia; and Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany
| | - Maria-Thereza Perez
- *Department of Clinical Sciences, Division of Ophthalmology, and NanoLund, Nanometer Structure Consortium, Lund University, Lund, Sweden; Graduate School of Cellular and Molecular Neuroscience, Center for Integrative Neuroscience (CIN), and Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany; Institute of Human Genetics, University of Regensburg, Regensburg, Germany; Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; **Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia; and Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany
| |
Collapse
|
143
|
Abstract
Astrocytes form borders (glia limitans) that separate neural from non-neural tissue along perivascular spaces, meninges and tissue lesions in the CNS. Transgenic loss-of-function studies reveal that astrocyte borders and scars serve as functional barriers that restrict the entry of inflammatory cells into CNS parenchyma in health and disease. Astrocytes also have powerful pro-inflammatory potential. Thus, astrocytes are emerging as pivotal regulators of CNS inflammatory responses. This Review discusses evidence that astrocytes have crucial roles in attracting and restricting CNS inflammation, with important implications for diverse CNS disorders.
Collapse
Affiliation(s)
- Michael V Sofroniew
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, California 90095, USA
| |
Collapse
|
144
|
Xiong Y, Zhang Y, Mahmood A, Chopp M. Investigational agents for treatment of traumatic brain injury. Expert Opin Investig Drugs 2015; 24:743-60. [PMID: 25727893 PMCID: PMC4433440 DOI: 10.1517/13543784.2015.1021919] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
INTRODUCTION Traumatic brain injury (TBI) is a major cause of death and disability worldwide. To date, there are no pharmacologic agents proven to improve outcomes from TBI because all the Phase III clinical trials in TBI have failed. Thus, there is a compelling need to develop treatments for TBI. AREAS COVERED The following article provides an overview of select cell-based and pharmacological therapies under early development for the treatment of TBI. These therapies seek to enhance cognitive and neurological functional recovery through neuroprotective and neurorestorative strategies. EXPERT OPINION TBI elicits both complex degenerative and regenerative tissue responses in the brain. TBI can lead to cognitive, behavioral, and motor deficits. Although numerous promising neuroprotective treatment options have emerged from preclinical studies that mainly target the lesion, translation of preclinical effective neuroprotective drugs to clinical trials has proven challenging. Accumulating evidence indicates that the mammalian brain has a significant, albeit limited, capacity for both structural and functional plasticity, as well as regeneration essential for spontaneous functional recovery after injury. A new therapeutic approach is to stimulate neurovascular remodeling by enhancing angiogenesis, neurogenesis, oligodendrogenesis, and axonal sprouting, which in concert, may improve neurological functional recovery after TBI.
Collapse
Affiliation(s)
- Ye Xiong
- Henry Ford Hospital, Department of Neurosurgery , Education and Research Building, Room 3096, 2799 West Grand Boulevard, Detroit, MI 48202 , USA +1 313 916 4743 ; +1 313 916 9855 ;
| | | | | | | |
Collapse
|
145
|
Teo JD, Morris MJ, Jones NM. Hypoxic postconditioning reduces microglial activation, astrocyte and caspase activity, and inflammatory markers after hypoxia-ischemia in the neonatal rat brain. Pediatr Res 2015; 77:757-64. [PMID: 25751571 DOI: 10.1038/pr.2015.47] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 11/12/2014] [Indexed: 11/09/2022]
Abstract
BACKGROUND Postconditioning (PostC) with mild hypoxia shortly after a neonatal hypoxic-ischemic (HI) brain injury can reduce brain damage, however, the mechanisms underlying this protection are not known. We hypothesize that hypoxic PostC reduces brain markers of glial activity, inflammation, and apoptosis following HI injury. METHODS Sprague Dawley rat pups were exposed to right common carotid artery occlusion and hypoxia (7% oxygen, 3 h) on postnatal day 7 and 24 h later, pups were exposed to hypoxic PostC (8% O2 for 1 h/day for 5 d) or kept at ambient conditions for the same duration. HI+N pups demonstrated ~10% loss in ipsilateral brain tissue which was rescued with HI+PostC. To investigate the cellular responses, markers of astrocytes, microglia, inflammation, and caspase 3 activity were examined using immunohistochemistry and enzyme-linked immunosorbent assay. RESULTS PostC reduced the area of astrocyte staining compared to HI+N. There was also a shift in microglial morphology toward a primed state in both PostC groups. Protein levels of interleukin-1β and caspase 3 were elevated in HI+N brains and reduced by PostC. CONCLUSION This is the first demonstration that PostC can reduce glial activity, inflammatory mediators, and cell death after a neonatal HI brain injury.
Collapse
Affiliation(s)
- Jonathan D Teo
- Department of Pharmacology, School of Medical Sciences, University of New South Wales, New South Wales, Australia
| | - Margaret J Morris
- Department of Pharmacology, School of Medical Sciences, University of New South Wales, New South Wales, Australia
| | - Nicole M Jones
- Department of Pharmacology, School of Medical Sciences, University of New South Wales, New South Wales, Australia
| |
Collapse
|
146
|
Shinjyo N, de Pablo Y, Pekny M, Pekna M. Complement Peptide C3a Promotes Astrocyte Survival in Response to Ischemic Stress. Mol Neurobiol 2015; 53:3076-3087. [DOI: 10.1007/s12035-015-9204-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 04/29/2015] [Indexed: 01/04/2023]
|
147
|
Choudhury GR, Ding S. Reactive astrocytes and therapeutic potential in focal ischemic stroke. Neurobiol Dis 2015; 85:234-244. [PMID: 25982835 DOI: 10.1016/j.nbd.2015.05.003] [Citation(s) in RCA: 175] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 03/26/2015] [Accepted: 05/08/2015] [Indexed: 12/17/2022] Open
Abstract
Astrocytes are specialized and the most abundant cell type in the central nervous system (CNS). They play important roles in the physiology of the brain. Astrocytes are also critically involved in many CNS disorders including focal ischemic stroke, the leading cause of brain injury and death in patients. One of the prominent pathological features of a focal ischemic stroke is reactive astrogliosis and glial scar formation. Reactive astrogliosis is accompanied with changes in morphology, proliferation, and gene expression in the reactive astrocytes. This study provides an overview of the most recent advances in astrocytic Ca(2+) signaling, spatial, and temporal dynamics of the morphology and proliferation of reactive astrocytes as well as signaling pathways involved in the reactive astrogliosis after ischemic stroke based on results from experimental studies performed in various animal models. This review also discusses the therapeutic potential of reactive astrocytes in focal ischemic stroke. As reactive astrocytes exhibit high plasticity, we suggest that modulation of local reactive astrocytes is a promising strategy for cell-based stroke therapy.
Collapse
Affiliation(s)
| | - Shinghua Ding
- Dalton Cardiovascular Research Center, Columbia, MO, USA; Department of Bioengineering, University of Missouri-Columbia, Columbia, MO 65211, USA.
| |
Collapse
|
148
|
Grégoire CA, Goldenstein BL, Floriddia EM, Barnabé-Heider F, Fernandes KJL. Endogenous neural stem cell responses to stroke and spinal cord injury. Glia 2015; 63:1469-82. [DOI: 10.1002/glia.22851] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 04/13/2015] [Indexed: 01/13/2023]
Affiliation(s)
- Catherine-Alexandra Grégoire
- Research Center of the University of Montreal Hospital (CRCHUM); Quebec Canada
- CNS Research Group (GRSNC), University of Montreal; Quebec Canada
- Department of Pathology and Cell Biology, Faculty of Medicine; Université De Montréal; Quebec Canada
| | - Brianna L. Goldenstein
- Research Center of the University of Montreal Hospital (CRCHUM); Quebec Canada
- CNS Research Group (GRSNC), University of Montreal; Quebec Canada
- Department of Neurosciences, Faculty of Medicine; Université De Montréal; Quebec Canada
| | | | | | - Karl J. L. Fernandes
- Research Center of the University of Montreal Hospital (CRCHUM); Quebec Canada
- CNS Research Group (GRSNC), University of Montreal; Quebec Canada
- Department of Neurosciences, Faculty of Medicine; Université De Montréal; Quebec Canada
| |
Collapse
|
149
|
Bruun DA, Cao Z, Inceoglu B, Vito ST, Austin AT, Hulsizer S, Hammock BD, Tancredi DJ, Rogawski MA, Pessah IN, Lein PJ. Combined treatment with diazepam and allopregnanolone reverses tetramethylenedisulfotetramine (TETS)-induced calcium dysregulation in cultured neurons and protects TETS-intoxicated mice against lethal seizures. Neuropharmacology 2015; 95:332-42. [PMID: 25882826 DOI: 10.1016/j.neuropharm.2015.03.035] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 03/15/2015] [Accepted: 03/31/2015] [Indexed: 01/09/2023]
Abstract
Tetramethylenedisulfotetramine (TETS) is a potent convulsant GABAA receptor blocker. Mice receiving a lethal dose of TETS (0.15 mg/kg i.p.) are rescued from death by a high dose of diazepam (5 mg/kg i.p.) administered shortly after the second clonic seizure (∼20 min post-TETS). However, this high dose of diazepam significantly impairs blood pressure and mobility, and does not prevent TETS-induced neuroinflammation in the brain. We previously demonstrated that TETS alters synchronous Ca(2+) oscillations in primary mouse hippocampal neuronal cell cultures and that pretreatment with the combination of diazepam and allopregnanolone at concentrations having negligible effects individually prevents TETS effects on intracellular Ca(2+) dynamics. Here, we show that treatment with diazepam and allopregnanolone (0.1 μM) 20 min after TETS challenge normalizes synchronous Ca(2+) oscillations when added in combination but not when added singly. Similarly, doses (0.03-0.1 mg/kg i.p.) of diazepam and allopregnanolone that provide minimal protection when administered singly to TETS intoxicated mice increase survival from 10% to 90% when given in combination either 10 min prior to TETS or following the second clonic seizure. This therapeutic combination has negligible effects on blood pressure or mobility. Combined treatment with diazepam and allopregnanolone also decreases TETS-induced microglial activation. Diazepam and allopregnanolone have distinct actions as positive allosteric modulators of GABAA receptors that in combination enhance survival and mitigate neuropathology following TETS intoxication without the adverse side effects associated with high dose benzodiazepines. Combination therapy with a benzodiazepine and neurosteroid represents a novel neurotherapeutic strategy with potentially broad application.
Collapse
Affiliation(s)
- Donald A Bruun
- Department of Molecular Biosciences, School of Veterinary Medicine, Davis, CA 95616, USA
| | - Zhengyu Cao
- Department of Molecular Biosciences, School of Veterinary Medicine, Davis, CA 95616, USA
| | - Bora Inceoglu
- Department of Entomology, College of Agricultural and Environmental Sciences, University of California, Davis, Davis, CA 95616, USA
| | - Stephen T Vito
- Department of Entomology, College of Agricultural and Environmental Sciences, University of California, Davis, Davis, CA 95616, USA
| | | | - Susan Hulsizer
- Department of Molecular Biosciences, School of Veterinary Medicine, Davis, CA 95616, USA
| | - Bruce D Hammock
- Department of Entomology, College of Agricultural and Environmental Sciences, University of California, Davis, Davis, CA 95616, USA; Comprehensive Cancer Center, Sacramento, CA 95817, USA
| | | | - Michael A Rogawski
- Department of Neurology, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Isaac N Pessah
- Department of Molecular Biosciences, School of Veterinary Medicine, Davis, CA 95616, USA
| | - Pamela J Lein
- Department of Molecular Biosciences, School of Veterinary Medicine, Davis, CA 95616, USA.
| |
Collapse
|
150
|
Huang Y, Tan S. Direct lineage conversion of astrocytes to induced neural stem cells or neurons. Neurosci Bull 2015; 31:357-67. [PMID: 25854678 DOI: 10.1007/s12264-014-1517-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 10/14/2014] [Indexed: 12/27/2022] Open
Abstract
Since the generation of induced pluripotent stem cells in 2006, cellular reprogramming has attracted increasing attention as a revolutionary strategy for cell replacement therapy. Recent advances have revealed that somatic cells can be directly converted into other mature cell types, which eliminates the risk of neoplasia and the generation of undesired cell types. Astrocytes become reactive and undergo proliferation, which hampers axon regeneration following injury, stroke, and neurodegenerative diseases. An emerging technique to directly reprogram astrocytes into induced neural stem cells (iNSCs) and induced neurons (iNs) by neural fate determinants brings potential hope to cell replacement therapy for the above neurological problems. Here, we discuss the development of direct reprogramming of various cell types into iNs and iNSCs, then detail astrocyte-derived iNSCs and iNs in vivo and in vitro. Finally, we highlight the unsolved challenges and opportunities for improvement.
Collapse
Affiliation(s)
- Yanhua Huang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282, China
| | | |
Collapse
|