1
|
Ma J, Zhang M, Fu P, Yin X, Chen Z. Chemokines play a role in nerve damage and neuroprotection in vascular dementia. IBRO Neurosci Rep 2024; 17:154-160. [PMID: 39206161 PMCID: PMC11350449 DOI: 10.1016/j.ibneur.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/30/2024] [Accepted: 08/03/2024] [Indexed: 09/04/2024] Open
Abstract
Various Chemotactic Factors (FCs) play different roles in neuronal injury in vascular dementia. CXCL5 and CCL11 exacerbate neurological injury by promoting inflammatory responses. CXCL12/SDF-1 and CX3CL1 play neuroprotective roles.CXCL13, XCL-1 and CCL2/ MCP-1 exacerbate neurological injury in the early stage, while exerting neuronal regeneration and neuroprotective effects in the chronic progressive phase. Chemokines often play an important role in the course of vascular dementia by regulating inflammatory responses, oxidative stress, and autophagy. Activation of microglia plays an important role in the regression of vascular dementia. Activated microglia M1 causes neuronal damage through the release of chemokines. And microglia M2 has anti-inflammatory effects and is involved in the repair of brain damage. Therefore, dynamic monitoring of various related FCs and understanding the relationship between FCs and microglia can help to understand and regulate the disease course progression of vascular dementia.At present, many scholars have confirmed in basic research that different subgroups of chemokines are closely related to vascular dementia. In clinical research, new immunotherapy methods that upregulate XCL-1 and drugs that regulate the activity of CCL2/CCR2 signaling pathways are being studied and promoted.
Collapse
Affiliation(s)
- Jinming Ma
- Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang,Jiangxi, 332000, China
| | - Manqing Zhang
- School of Basic Medicine, Jiujiang University, Jiujiang, Jiangxi, 332000, China
| | - Peijie Fu
- Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang,Jiangxi, 332000, China
| | - Xiaoping Yin
- Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang,Jiangxi, 332000, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi,332000, China
| | - Zhiying Chen
- Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang,Jiangxi, 332000, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi,332000, China
| |
Collapse
|
2
|
Barrientos-Bonilla AA, Pensado-Guevara PB, Puga-Olguín A, Nadella R, Sánchez-García ADC, Zavala-Flores LM, Villanueva-Olivo A, Cibrián-Llanderal IT, Rovirosa-Hernández MDJ, Hernandez-Baltazar D. BrdU does not induce hepatocellular damage in experimental Wistar rats. Acta Histochem 2024; 126:152117. [PMID: 38016413 DOI: 10.1016/j.acthis.2023.152117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 11/03/2023] [Accepted: 11/13/2023] [Indexed: 11/30/2023]
Abstract
Bromodeoxyuridine (BrdU) is used in studies related to cell proliferation and neurogenesis. The multiple intraperitoneal injections of this molecule could favor liver function profile changes. In this study, we evaluate the systemic and hepatocellular impact of BrdU in male adult Wistar rats in 30 %-partial hepatectomy (PHx) model. The rats received BrdU 50 mg/Kg by intraperitoneal injection at 0.5, 1, 2, 3, 6, 9 and 16 days after 30 %-PH. The rats were distributed into four groups as follows, control, sham, PHx/BrdU(-) and PHx/BrdU(+). On day 16, we evaluated hepatocellular nuclei and analyzed histopathological features by haematoxylin-eosin stain and apoptotic profile was qualified by caspase-3 presence. The systemic effect was evaluated by liver markers such as alanine transferase (ALT), aspartate aminotransferase (AST), lactate dehydrogenase (LDH), alkaline phosphatase (AP), bilirubin, total proteins and serum albumin content. The statistical analysis consisted of a student t-test and one-way ANOVA. BrdU did not induce apoptosis or hepatocellular damage in male rats. Multiple administrations of BrdU in male rats did not induce significant decrease body weight, but increased serum ALT and LDH levels were found. Our results show that the BrdU does not produce hepatocellular damage.
Collapse
Affiliation(s)
| | | | - Abraham Puga-Olguín
- Unidad de Salud Integrativa, Centro de EcoAlfabetización y Diálogo de Saberes, Universidad Veracruzana, Xalapa, Veracruz, Mexico
| | | | | | | | - Arnulfo Villanueva-Olivo
- Departamento de Histología. Facultad de Medicina. Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, Mexico
| | | | | | - Daniel Hernandez-Baltazar
- Instituto de Neuroetología, Universidad Veracruzana, Xalapa, Veracruz, Mexico; Investigadoras e investigadores por México CONAHCyT-Instituto de Neuroetología, Universidad Veracruzana, Mexico.
| |
Collapse
|
3
|
Sawada K, Kamiya S, Kobayashi T. Neonatal Exposure to Lipopolysaccharide Promotes Neurogenesis of Subventricular Zone Progenitors in the Developing Neocortex of Ferrets. Int J Mol Sci 2023; 24:14962. [PMID: 37834410 PMCID: PMC10573966 DOI: 10.3390/ijms241914962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 09/29/2023] [Accepted: 10/03/2023] [Indexed: 10/15/2023] Open
Abstract
Lipopolysaccharide (LPS) is a natural agonist of toll-like receptor 4 that serves a role in innate immunity. The current study evaluated the LPS-mediated regulation of neurogenesis in the subventricular zone (SVZ) progenitors, that is, the basal radial glia and intermediate progenitors (IPs), in ferrets. Ferret pups were subcutaneously injected with LPS (500 μg/g of body weight) on postnatal days (PDs) 6 and 7. Furthermore, 5-ethynyl-2'-deoxyuridine (EdU) and 5-bromo-2'-deoxyuridine (BrdU) were administered on PDs 5 and 7, respectively, to label the post-proliferative and proliferating cells in the inner SVZ (iSVZ) and outer SVZ (oSVZ). A significantly higher density of BrdU single-labeled proliferating cells was observed in the iSVZ of LPS-exposed ferrets than in controls but not in post-proliferative EdU single-labeled and EdU/BrdU double-labeled self-renewing cells. BrdU single-labeled cells exhibited a lower proportion of Tbr2 immunostaining in LPS-exposed ferrets (22.2%) than in controls (42.6%) and a higher proportion of Ctip2 immunostaining in LPS-exposed ferrets (22.2%) than in controls (8.6%). The present findings revealed that LPS modified the neurogenesis of SVZ progenitors. Neonatal LPS exposure facilitates the proliferation of SVZ progenitors, followed by the differentiation of Tbr2-expressing IPs into Ctip2-expressing immature neurons.
Collapse
Affiliation(s)
- Kazuhiko Sawada
- Department of Nutrition, Faculty of Medical and Health Sciences, Tsukuba International University, Tsuchiura 300-0051, Japan
| | - Shiori Kamiya
- Department of Regulation Biology, Faculty of Science, Saitama University, Saitama 338-8570, Japan; (S.K.); (T.K.)
| | - Tetsuya Kobayashi
- Department of Regulation Biology, Faculty of Science, Saitama University, Saitama 338-8570, Japan; (S.K.); (T.K.)
| |
Collapse
|
4
|
Shabani Z, Soltani Zangbar H, Nasrolahi A. Cerebral dopamine neurotrophic factor increases proliferation, Migration and differentiation of subventricular zone neuroblasts in photothrombotic stroke model of mouse. J Stroke Cerebrovasc Dis 2022; 31:106725. [PMID: 36116218 DOI: 10.1016/j.jstrokecerebrovasdis.2022.106725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 07/26/2022] [Accepted: 08/08/2022] [Indexed: 10/31/2022] Open
Abstract
BACKGROUND Cerebral ischemic stroke can induce the proliferation of subventricular zone (SVZ) neural stem cells (NSCs) in the adult brain. However, this reparative process is restricted because of NSCs' death shortly after injury or disability of them to reach the infarct boundary. In the present study, we investigated the ability of cerebral dopamine neurotrophic factor (CDNF) on the attraction of SVZ-resident NSCs toward the lesioned area and neurological recovery in a photothrombotic (PT) stroke model of mice METHODS: The mice were assigned to three groups stroke, stroke+phosphate buffered saline (PBS), and stroke+CDNF. Migration of SVZ NSCs were evaluated by BrdU/doublecortin (DCX) double immunofluorescence method on days 7 and 14 and their differentiation were evaluated by BrdU/ Neuronal Nuclei (NeuN) double immunofluorescence method 28 days after intra-SVZ CDNF injection. Serial coronal sections were stained with cresyl violet to detect the infarct volume and a modified neurological severity score (mNSS) was performed to assess the neurological performance RESULTS: Injection of CDNF increased the proliferation of SVZ NSCs and the number of DCX-expressing neuroblasts migrated from the SVZ toward the ischemic site. It also enhanced the differentiation of migrated neuroblasts into the mature neurons in the lesioned site. Along with this, the infarct volume was significantly decreased and the neurological performance was improved as compared to other groups CONCLUSION: These results demonstrate that CDNF is capable of enhancing the proliferation of NSCs residing in the SVZ and their migration toward the ischemia region and finally, differentiation of them in stroke mice, concomitantly decreased infarct volume and improved neurological abilities were revealed.
Collapse
Affiliation(s)
- Zahra Shabani
- Center for Cerebrovascular Research, University of California, San Francisco, California, USA; Infectious Ophthalmologic Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hamid Soltani Zangbar
- Department of Neuroscience and Cognition, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ava Nasrolahi
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
5
|
Grade S, Thomas J, Zarb Y, Thorwirth M, Conzelmann KK, Hauck SM, Götz M. Brain injury environment critically influences the connectivity of transplanted neurons. SCIENCE ADVANCES 2022; 8:eabg9445. [PMID: 35687687 PMCID: PMC9187233 DOI: 10.1126/sciadv.abg9445] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Cell transplantation is a promising approach for the reconstruction of neuronal circuits after brain damage. Transplanted neurons integrate with remarkable specificity into circuitries of the mouse cerebral cortex affected by neuronal ablation. However, it remains unclear how neurons perform in a local environment undergoing reactive gliosis, inflammation, macrophage infiltration, and scar formation, as in traumatic brain injury (TBI). To elucidate this, we transplanted cells from the embryonic mouse cerebral cortex into TBI-injured, inflamed-only, or intact cortex of adult mice. Brain-wide quantitative monosynaptic rabies virus (RABV) tracing unraveled graft inputs from correct regions across the brain in all conditions, with pronounced quantitative differences: scarce in intact and inflamed brain versus exuberant after TBI. In the latter, the initial overshoot is followed by pruning, with only a few input neurons persisting at 3 months. Proteomic profiling identifies candidate molecules for regulation of the synaptic yield, a pivotal parameter to tailor for functional restoration of neuronal circuits.
Collapse
Affiliation(s)
- Sofia Grade
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians University Munich, 82152 Planegg-Martinsried, Germany
- Institute of Stem Cell Research, Helmholtz Center Munich, German Center for Environmental Health, 82152 Planegg-Martinsried, Germany
- Corresponding author. (S.G.); (S.M.H.); (M.G.)
| | - Judith Thomas
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians University Munich, 82152 Planegg-Martinsried, Germany
- Institute of Stem Cell Research, Helmholtz Center Munich, German Center for Environmental Health, 82152 Planegg-Martinsried, Germany
- Graduate School of Systemic Neuroscience, Ludwig-Maximilians University Munich, 82152 Planegg-Martinsried, Germany
| | - Yvette Zarb
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians University Munich, 82152 Planegg-Martinsried, Germany
- Institute of Stem Cell Research, Helmholtz Center Munich, German Center for Environmental Health, 82152 Planegg-Martinsried, Germany
| | - Manja Thorwirth
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians University Munich, 82152 Planegg-Martinsried, Germany
- Institute of Stem Cell Research, Helmholtz Center Munich, German Center for Environmental Health, 82152 Planegg-Martinsried, Germany
| | - Karl-Klaus Conzelmann
- Max von Pettenkofer Institute Virology, Medical Faculty and Gene Center, Ludwig-Maximilians University Munich, 81377 Munich, Germany
| | - Stefanie M. Hauck
- Research Unit Protein Science and Metabolomics and Proteomics Core, Helmholtz Center Munich, German Center for Environmental Health, 85764 Neuherberg, Germany
- Corresponding author. (S.G.); (S.M.H.); (M.G.)
| | - Magdalena Götz
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians University Munich, 82152 Planegg-Martinsried, Germany
- Institute of Stem Cell Research, Helmholtz Center Munich, German Center for Environmental Health, 82152 Planegg-Martinsried, Germany
- SYNERGY, Excellence Cluster of Systems Neurology, Biomedical Center, Ludwig-Maximilians University Munich, 82152 Planegg-Martinsried, Germany
- Corresponding author. (S.G.); (S.M.H.); (M.G.)
| |
Collapse
|
6
|
Prophylactic Zinc Administration Combined with Swimming Exercise Prevents Cognitive-Emotional Disturbances and Tissue Injury following a Transient Hypoxic-Ischemic Insult in the Rat. Behav Neurol 2022; 2022:5388944. [PMID: 35637877 PMCID: PMC9146809 DOI: 10.1155/2022/5388944] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 03/04/2022] [Accepted: 04/01/2022] [Indexed: 11/17/2022] Open
Abstract
Exercise performance and zinc administration individually yield a protective effect on various neurodegenerative models, including ischemic brain injury. Therefore, this work was aimed at evaluating the combined effect of subacute prophylactic zinc administration and swimming exercise in a transient cerebral ischemia model. The prophylactic zinc administration (2.5 mg/kg of body weight) was provided every 24 h for four days before a 30 min common carotid artery occlusion (CCAO), and 24 h after reperfusion, the rats were subjected to swimming exercise in the Morris Water Maze (MWM). Learning was evaluated daily for five days, and memory on day 12 postreperfusion; anxiety or depression-like behavior was measured by the elevated plus maze and the motor activity by open-field test. Nitrites, lipid peroxidation, and the activity of superoxide dismutase (SOD) and catalase (CAT) were assessed in the temporoparietal cortex and hippocampus. The three nitric oxide (NO) synthase isoforms, chemokines, and their receptor levels were measured by ELISA. Nissl staining evaluated hippocampus cytoarchitecture and Iba-1 immunohistochemistry activated the microglia. Swimming exercise alone could not prevent ischemic damage but, combined with prophylactic zinc administration, reversed the cognitive deficit, decreased NOS and chemokine levels, prevented tissue damage, and increased Iba-1 (+) cell number. These results suggest that the subacute prophylactic zinc administration combined with swimming exercise, but not the individual treatment, prevents the ischemic damage on day 12 postreperfusion in the transient ischemia model.
Collapse
|
7
|
Drug repurposing for stroke intervention. Drug Discov Today 2022; 27:1974-1982. [DOI: 10.1016/j.drudis.2022.03.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 02/07/2023]
|
8
|
Ye X, Peng X, Song Q, Zeng T, Xiong X, Huang Y, Cai X, Zhang C, Wang C, Wang B. Borneol-modified tanshinone IIA liposome improves cerebral ischemia reperfusion injury by suppressing NF-κB and ICAM-1 expression. Drug Dev Ind Pharm 2021; 47:609-617. [PMID: 33834937 DOI: 10.1080/03639045.2021.1908331] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To investigate the metabolism and brain tissue distribution of borneol-modified tanshinone IIA liposome (BO-TA-Lip) and its effect on NF-κB and ICAM-1 in cerebral ischemia reperfusion rats, thereby exploring the ameliorative mechanism of BO-TA-Lip on ischemic encephalopathy. METHODS Particle size, entrapment efficiency, drug loading were measured to evaluate the preparation comprehensively. Metabolism and brain tissue distributions in vivo were measured by HPLC, and the pharmacokinetic parameters were calculated. In addition, 24 SD rats were randomly divided into sham, model, STS (sodium tanshinone IIA sulfonate, 30 mg/kg) and BO-TA-Lip groups (44 mg/kg). The middle cerebral artery occlusion (MCAO) rats were constructed with thread embolism method. Neurological deficits were scored using Zea Longa scoring standard. TTC and HE staining were used for the cerebral infarction and histopathological examination, respectively. The protein expression was examined by immunohistochemistry and Western blot. RESULTS The average particle size, encapsulation efficiency and drug loading of BO-TA-Lip were (135.33 ± 7.25) nm, (85.95 ± 3.20)% and (4.06 ± 0.31)%, respectively. Both in the pharmacokinetic analysis of plasma and brain tissue, in BO-TA-Lip group, the peak concentration and the area under the curve increased, and the clearance rate decreased. The neurological deficit scores and infarct area of the BO-TA-Lip group were significantly lower than that of the model and STS groups. Besides, BO-TA-Lip reduced the protein expression of NF-κB, ICAM-1, IL-1β, TNF-α and IL-6 in the brain tissue. CONCLUSION BO-TA-Lip had higher bioavailability and better absorption in brain tissue, and could improve cerebral ischemia reperfusion injury, which might be related to the inhibitory effect of BO-TA-Lip in expression of NF-κB and ICAM-1.
Collapse
Affiliation(s)
- Xiaoli Ye
- Department of Clinical Pharmacy, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xueying Peng
- Department of Clinical Pharmacy, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qing Song
- Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Taohui Zeng
- First Affiliated Hospital, Gannan Medical College, Ganzhou, China
| | | | - Yuye Huang
- The Affiliated Cangnan Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xinjun Cai
- Zhejiang Chinese Medicine and Western Medicine Integrated Hospital, Hangzhou, Zhejiang, China
| | - Chao Zhang
- Hangzhou Lin'an district People's Hospital, Hangzhou, Zhejiang, China
| | - Congyao Wang
- The First People's Hospital of Xiaoshan District, Hangzhou, Zhejiang, China
| | - Binhui Wang
- Municipal Hospital Affiliated to Medical School of Taizhou University, Taizhou, Zhejiang, China
| |
Collapse
|
9
|
Dail ME, Brino MLM, Chambers JE. Effects of novel brain-penetrating oxime acetylcholinesterase reactivators on sarin surrogate-induced changes in rat brain gene expression. J Biochem Mol Toxicol 2021; 35:1-10. [PMID: 33682265 DOI: 10.1002/jbt.22755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 01/28/2021] [Accepted: 02/24/2021] [Indexed: 11/11/2022]
Abstract
Past assassinations and terrorist attacks demonstrate the need for a more effective antidote against nerve agents and other organophosphates (OP) that cause brain damage through inhibition of acetylcholinesterase (AChE). Our lab has invented a platform of phenoxyalkyl pyridinium oximes (US patent 9,277,937) that demonstrate the ability to cross the blood-brain barrier in in vivo rat tests with a sarin surrogate nitrophenyl isopropyl methylphosphonate (NIMP) and provide evidence of brain penetration by reducing cessation time of seizure-like behaviors, accumulation of glial fibrillary acidic protein (GFAP), and hippocampal neuropathology, as opposed to the currently approved oxime, 2-pyridine aldoxime methyl chloride (2-PAM). Using two of the novel oximes (Oximes 1 and 20), this project examined whether gene expression changes might help explain this protection. Expression changes in the piriform cortex were examined using polymerase chain reaction arrays for inflammatory cytokines and receptors. The hippocampus was examined via quantitative polymerase chain reaction for the expression of immediate-early genes involved in brain repair (Bdnf), increasing neurotoxicity (Fos), and apoptosis control (Jdp2, Bcl2l1, Bcl2l11). In the piriform cortex, NIMP significantly stimulated expression for the macrophage inflammatory proteins CCL4, IL-1A, and IL-1B. Oxime 20 by itself elicited the most changes. When it was given therapeutically post-NIMP, the largest change occurred: a 310-fold repression of the inflammatory cytokine, CCL12. In the hippocampus, NIMP increased the expression of the neurotoxicity marker Fos and decreased the expression of neuroprotective Bdnf and antiapoptotic Bcl2l1. Compared with 2-PAM, Oxime 20 stimulated Bcl2l1 expression more and returned expression closer to the vehicle control values.
Collapse
Affiliation(s)
- Mary E Dail
- Department of Comparative Biomedical Sciences, Center for Environmental Health Sciences, Mississippi State University, College of Veterinary Medicine, Mississippi State, United States, USA
| | - Meghan L M Brino
- Department of Comparative Biomedical Sciences, Center for Environmental Health Sciences, Mississippi State University, College of Veterinary Medicine, Mississippi State, United States, USA
| | - Janice E Chambers
- Department of Comparative Biomedical Sciences, Center for Environmental Health Sciences, Mississippi State University, College of Veterinary Medicine, Mississippi State, United States, USA
| |
Collapse
|
10
|
Dos Santos IRC, Dias MNC, Gomes-Leal W. Microglial activation and adult neurogenesis after brain stroke. Neural Regen Res 2021; 16:456-459. [PMID: 32985465 PMCID: PMC7996005 DOI: 10.4103/1673-5374.291383] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The discovery that new neurons are produced in some regions of the adult mammalian brain is a paradigm-shift in neuroscience research. These new-born cells are produced from neuroprogenitors mainly in the subventricular zone at the margin of the lateral ventricle, subgranular zone in the hippocampal dentate gyrus and in the striatum, a component of the basal ganglia, even in humans. In the human hippocampus, neuroblasts are produced even in elderlies. The regulation of adult neurogenesis is a complex phenomenon involving a multitude of molecules, neurotransmitters and soluble factors released by different sources including glial cells. Microglia, the resident macrophages of the central nervous system, are considered to play an important role on the regulation of adult neurogenesis both in physiological and pathological conditions. Following stroke and other acute neural disorders, there is an increase in the numbers of neuroblast production in the neurogenic niches. Microglial activation is believed to display both beneficial and detrimental role on adult neurogenesis after stroke, depending on the activation level and brain location. In this article, we review the scientific evidence addressing the role of microglial activation on adult neurogenesis after ischemia. A comprehensive understanding of the microglial role after stroke and other neural disorders it is an important step for development of future therapies based on manipulation of adult neurogenesis.
Collapse
Affiliation(s)
- Ijair R C Dos Santos
- Laboratory of Experimental Neuroprotection and Neuroregeneration, Institute of Biological Sciences, Federal University of Pará-Brazil, Belém-Pará, Brazil
| | - Michelle Nerissa C Dias
- Laboratory of Experimental Neuroprotection and Neuroregeneration, Institute of Biological Sciences, Federal University of Pará-Brazil, Belém-Pará, Brazil
| | - Walace Gomes-Leal
- Laboratory of Experimental Neuroprotection and Neuroregeneration, Institute of Biological Sciences, Federal University of Pará-Brazil, Belém-Pará, Brazil
| |
Collapse
|
11
|
Shen Y, Zhang Y, Chen L, Du J, Bao H, Xing Y, Cai M, Si Y. Chemokine CXCL13 acts via CXCR5-ERK signaling in hippocampus to induce perioperative neurocognitive disorders in surgically treated mice. J Neuroinflammation 2020; 17:335. [PMID: 33161894 PMCID: PMC7648984 DOI: 10.1186/s12974-020-02013-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 10/28/2020] [Indexed: 12/19/2022] Open
Abstract
Background Perioperative neurocognitive disorders (PNDs) occur frequently after surgery and worsen patient outcome. How C-X-C motif chemokine (CXCL) 13 and its sole receptor CXCR5 contribute to PNDs remains poorly understood. Methods A PND model was created in adult male C57BL/6J and CXCR5−/− mice by exploratory laparotomy. Mice were pretreated via intracerebroventricular injection with recombinant CXCL13, short hairpin RNA against CXCL13 or a scrambled control RNA, or ERK inhibitor PD98059. Then surgery was performed to induce PNDs, and animals were assessed in the Barnes maze trial followed by a fear-conditioning test. Expression of CXCL13, CXCR5, and ERK in hippocampus was examined using Western blot, quantitative PCR, and immunohistochemistry. Levels of interleukin-1 beta (IL-1β) and tumor necrosis factor alpha (TNF-α) in hippocampus were assessed by Western blot. Results Surgery impaired learning and memory, and it increased expression of CXCL13 and CXCR5 in the hippocampus. CXCL13 knockdown partially reversed the effects of surgery on CXCR5 and cognitive dysfunction. CXCR5 knockout led to similar cognitive outcomes as CXCL13 knockdown, and it repressed surgery-induced activation of ERK and production of IL-1β and TNF-α in hippocampus. Recombinant CXCL13 induced cognitive deficits and increased the expression of phospho-ERK as well as IL-1β and TNF-α in hippocampus of wild-type mice, but not CXCR5−/− mice. PD98059 partially blocked CXCL13-induced cognitive dysfunction as well as production of IL-1β and TNF-α. Conclusions CXCL13-induced activation of CXCR5 may contribute to PNDs by triggering ERK-mediated production of pro-inflammatory cytokines in hippocampus.
Collapse
Affiliation(s)
- Yanan Shen
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, People's Republic of China
| | - Yuan Zhang
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, People's Republic of China
| | - Lihai Chen
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, People's Republic of China
| | - Jiayue Du
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, People's Republic of China
| | - Hongguang Bao
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, People's Republic of China
| | - Yan Xing
- Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, School of Mechanical Engineering, Southeast University, Nanjing, 211118, People's Republic of China
| | - Mengmeng Cai
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, People's Republic of China
| | - Yanna Si
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, People's Republic of China.
| |
Collapse
|
12
|
Guo T, Liu Y, Ren X, Wang W, Liu H. Promoting Role of Long Non-Coding RNA Small Nucleolar RNA Host Gene 15 (SNHG15) in Neuronal Injury Following Ischemic Stroke via the MicroRNA-18a/CXC Chemokine Ligand 13 (CXCL13)/ERK/MEK Axis. Med Sci Monit 2020; 26:e923610. [PMID: 32862188 PMCID: PMC7480088 DOI: 10.12659/msm.923610] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Long-non-coding RNA (lncRNA) SNHG15 has been reported to be an aberrantly expressed lncRNA in patients with ischemic stroke, but its role in neuronal injury following ischemic stroke remains unclear. We hypothesized that this lncRNA is associated with the pathogenesis of ischemic stroke. Material/Methods A mouse model of ischemic stroke was established by middle cerebral artery occlusion (MCAO). A neurogenic mouse cell line Neuro-2a (N2a) was subjected to oxygen-glucose deprivation (OGD) for in vitro experiments. Expression of SNHG15, microRNA-18a (miR-18a), and CXCL13 in mouse brain and in OGD-treated N2a cells was determined. Altered expression of SNHG15 and miR-18a was introduced to detect their roles in N2a cell viability and apoptosis. Targeting relationships between miR-18a and SNHG15 or CXCL13 were validated by luciferase assays. Cells were treated with the ERK/MEK antagonist U0126 to assess the role of the ERK/MEK signaling pathway in N2a cell growth. Results SNHG15 and CXCL13 were overexpressed and miR-18a was underexpressed in MCAO-induced mice and OGD-treated N2a cells. Silencing of SNHG15 or overexpression of miR-18a promoted cell viability, while decreased cell apoptosis induced by OGD; however, subsequent disruption of the ERK/MEK signaling pathway reversed these effects. SNHG15 was found to bind to miR-18a, which could further target CXCL13. Conclusions Silencing of SNHG15 led to CXCL13 upregulation through sequestering miR-18a and the following ERK/MEK activation, thus enhancing viability while reducing apoptosis of N2a cells. SNHG15 may serve as a novel target for ischemic stroke treatment.
Collapse
Affiliation(s)
- Tiezhu Guo
- Department of Neurosurgery, Heji Hospital Affiliated with Changzhi Medical College, Changzhi, Shanxi, China (mainland)
| | - Yueting Liu
- Department of Neurosurgery, The First Hospital Affiliated with Shanxi Medical University, Taiyuan, Shanxi, China (mainland)
| | - Xinliang Ren
- Department of Neurosurgery, Heji Hospital Affiliated with Changzhi Medical College, Changzhi, Shanxi, China (mainland)
| | - Wei Wang
- Department of Neurosurgery, Heji Hospital Affiliated with Changzhi Medical College, Changzhi, Shanxi, China (mainland)
| | - Hanrui Liu
- Department of Neurology, Heji Hospital Affiliated with Changzhi Medical College, Changzhi, Shanxi, China (mainland)
| |
Collapse
|
13
|
Xu D, Li F, Xue G, Hou K, Fang W, Li Y. Effect of Wnt signaling pathway on neurogenesis after cerebral ischemia and its therapeutic potential. Brain Res Bull 2020; 164:1-13. [PMID: 32763283 DOI: 10.1016/j.brainresbull.2020.07.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 12/08/2019] [Accepted: 07/08/2020] [Indexed: 02/07/2023]
Abstract
Neurogenesis process in the chronic phase of ischemic stroke has become the focus of research on stroke treatment recently, mainly through the activation of related pathways to increase the differentiation of neural stem cells (NSCs) in the brain sub-ventricular zone (SVZ) and subgranular zone (SGZ) of hippocampal dentate gyrus (DG) areas into neurons, promoting neurogenesis. While there is still debate about the longevity of active adult neurogenesis in humans, the SVZ and SGZ have the capacity to upregulate neurogenesis in response to cerebral ischemia, which opens discussion about potential treatment strategies to harness this neuronal regenerative response. Wnt signaling pathway is one of the most important approaches potentially targeting on neurogenesis after cerebral ischemia, appropriate activation of which in NSCs may help to improve the sequelae of cerebral ischemia. Various therapeutic approaches are explored on preclinical stage to target endogenous neurogenesis induced by Wnt signaling after stroke onset. This article describes the composition of Wnt signaling pathway and the process of neurogenesis after cerebral ischemia, and emphatically introduces the recent studies on the mechanisms of this pathway for post-stroke neurogenesis and the therapeutic possibility of activating the pathway to improve neurogenesis after stroke.
Collapse
Affiliation(s)
- Dan Xu
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, China.
| | - Fengyang Li
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, China.
| | - Gou Xue
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, China.
| | - Kai Hou
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, China.
| | - Weirong Fang
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, China.
| | - Yunman Li
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
14
|
Inflammation Induced by Natural Neuronal Death and LPS Regulates Neural Progenitor Cell Proliferation in the Healthy Adult Brain. eNeuro 2020; 7:ENEURO.0023-20.2020. [PMID: 32424053 PMCID: PMC7333977 DOI: 10.1523/eneuro.0023-20.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 05/11/2020] [Accepted: 05/12/2020] [Indexed: 12/28/2022] Open
Abstract
Inflammation is typically considered a negative response to injury or insult; however, recent advances demonstrate that inflammatory cells regulate development, plasticity, and homeostasis through anticytotoxic, progenerative responses. Here, we extend analyses of neuroinflammation to natural neurodegenerative and homeostatic states by exploiting seasonal plasticity in cytoarchitecture of the avian telencephalic song control nucleus, high vocal center [HVC (proper name)], in the songbird Gambel's white-crowned sparrow (Zonotrichia leucophrys gambelii). We report that local injection of the endotoxin lipopolysaccharide into HVC of birds in both breeding (high circulating testosterone level) and nonbreeding (low circulating testosterone level) conditions increased neural progenitor cell proliferation in the nearby but distinct ventricular zone. Additionally, we found that oral administration of the anti-inflammatory drug minocycline during seasonal regression of HVC reduced microglia activation in HVC and prevented the normal proliferative response in the ventricular zone to apoptosis in HVC. Our results suggest that local neuroinflammation positively regulates neural progenitor cell proliferation and, in turn, contributes to the previously described repatterning of HVC cytoarchitecture following seasonally induced neuronal loss.
Collapse
|
15
|
Fritze J, Ginisty A, McDonald R, Quist E, Stamp E, Monni E, Dhapola P, Lang S, Ahlenius H. Loss of Cxcr5 alters neuroblast proliferation and migration in the aged brain. Stem Cells 2020; 38:1175-1187. [PMID: 32469107 DOI: 10.1002/stem.3207] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 04/06/2020] [Accepted: 04/23/2020] [Indexed: 11/09/2022]
Abstract
Neurogenesis, the production of new neurons from neural stem cells, dramatically decreases during aging concomitantly with increased inflammation both systemically and in the brain. However, the precise role of inflammation and whether local or systemic factors drive the neurogenic decline during aging is poorly understood. Here, we identify CXCR5/5/CXCL13 signaling as a novel regulator of neurogenesis in the aged brain. The chemokine Cxcl13 was found to be upregulated in the brain during aging. Loss of its receptor, Cxcr5, led to increased proliferation and decreased numbers of neuroblasts in the aged subventricular zone (SVZ), together with accumulation of neuroblasts in the rostral migratory stream and olfactory bulb (OB), without increasing the amount of new mature neurons in the OB. The effect on proliferation and migration was specific to neuroblasts and likely mediated through increased levels of systemic IL-6 and local Cxcl12 expression in the SVZ. Our study raises the possibility of a new mechanism by which interplay between systemic and local alterations in inflammation regulates neurogenesis during aging.
Collapse
Affiliation(s)
- Jonas Fritze
- Faculty of Medicine, Department of Clinical Sciences and Neurology, Stem Cells, Aging and Neurodegeneration Group, Lund University, Lund, Sweden.,Lund Stem Cell Center, Lund, Sweden
| | - Aurélie Ginisty
- Faculty of Medicine, Department of Clinical Sciences and Neurology, Stem Cells, Aging and Neurodegeneration Group, Lund University, Lund, Sweden.,Lund Stem Cell Center, Lund, Sweden
| | - Rebecca McDonald
- Faculty of Medicine, Department of Clinical Sciences and Neurology, Stem Cells, Aging and Neurodegeneration Group, Lund University, Lund, Sweden.,Lund Stem Cell Center, Lund, Sweden
| | - Ella Quist
- Faculty of Medicine, Department of Clinical Sciences and Neurology, Stem Cells, Aging and Neurodegeneration Group, Lund University, Lund, Sweden.,Lund Stem Cell Center, Lund, Sweden
| | - Eleanor Stamp
- Faculty of Medicine, Department of Clinical Sciences and Neurology, Stem Cells, Aging and Neurodegeneration Group, Lund University, Lund, Sweden.,Lund Stem Cell Center, Lund, Sweden
| | - Emanuela Monni
- Lund Stem Cell Center, Lund, Sweden.,Faculty of Medicine, Department of Clinical Sciences and Neurology, Laboratory of Stem Cells and Restorative Neurology, Lund University, Lund, Sweden
| | - Parashar Dhapola
- Lund Stem Cell Center, Lund, Sweden.,Faculty of Medicine, Division of Molecular Hematology, Stem Cells and Leukemia Group, Lund University, Lund, Sweden
| | - Stefan Lang
- Lund Stem Cell Center, Lund, Sweden.,Faculty of Medicine, Division of Molecular Hematology, Computational Genomics Group, Lund University, Lund, Sweden
| | - Henrik Ahlenius
- Faculty of Medicine, Department of Clinical Sciences and Neurology, Stem Cells, Aging and Neurodegeneration Group, Lund University, Lund, Sweden.,Lund Stem Cell Center, Lund, Sweden
| |
Collapse
|
16
|
CXCR5 Knockdown Attenuates Hippocampal Neurogenesis Deficits and Cognitive Impairment in a Mouse Model of Sepsis-associated Encephalopathy. Neuroscience 2020; 433:212-220. [DOI: 10.1016/j.neuroscience.2020.03.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 10/28/2019] [Accepted: 03/08/2020] [Indexed: 01/21/2023]
|
17
|
Fisch U, Brégère C, Geier F, Chicha L, Guzman R. Neonatal hypoxia-ischemia in rat elicits a region-specific neurotrophic response in SVZ microglia. J Neuroinflammation 2020; 17:26. [PMID: 31954397 PMCID: PMC6969423 DOI: 10.1186/s12974-020-1706-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 01/08/2020] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Recent findings describe microglia as modulators of neurogenesis in the subventricular zone (SVZ). SVZ microglia in the adult rat are thought to adopt a neurotrophic phenotype after ischemic stroke. Early postnatal microglia are endogenously activated and may therefore exhibit an increased sensitivity to neonatal hypoxia-ischemia (HI). The goal of this study was to investigate the impact of cortico-striatal HI on the microglial phenotype, function, and gene expression in the early postnatal SVZ. METHODS Postnatal day (P)7 rats underwent sham or right-hemispheric HI surgery. Microglia in the SVZ, the uninjured cortex, and corpus callosum were immunohistochemically analyzed at P10, P20, and P40. The transcriptome of microdissected SVZ and cortical microglia was analyzed at P10 and P20, and the effect of P10 SVZ microglia on neurosphere generation in vitro was studied. RESULTS The microglial response to HI was region-specific. In the SVZ, a microglial accumulation, prolonged activation and phagocytosis was noted that was not observed in the cortex and corpus callosum. The transcriptome of SVZ microglia and cortical microglia were distinct, and after HI, SVZ microglia concurrently upregulated pro- and anti-inflammatory as well as neurotrophic genes. In vitro, microglia isolated from the SVZ supported neurosphere generation in a concentration-dependent manner. CONCLUSIONS Microglia are an inherent cellular component of the early postnatal SVZ and undergo developmental changes that are affected on many aspects by neonatal HI injury. Our results demonstrate that early postnatal SVZ microglia are sensitive to HI injury and display a long-lasting region-specific response including neurotrophic features.
Collapse
Affiliation(s)
- Urs Fisch
- Department of Neurology, University Hospital Basel, University Basel, Basel, Switzerland.
- Brain ischemia and regeneration, Department of Biomedicine, University Hospital Basel, University Basel, Basel, Switzerland.
| | - Catherine Brégère
- Brain ischemia and regeneration, Department of Biomedicine, University Hospital Basel, University Basel, Basel, Switzerland
| | - Florian Geier
- Bioinformatics Core Facility, Department of Biomedicine, University Basel, Basel, Switzerland
- Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Laurie Chicha
- Brain ischemia and regeneration, Department of Biomedicine, University Hospital Basel, University Basel, Basel, Switzerland
| | - Raphael Guzman
- Brain ischemia and regeneration, Department of Biomedicine, University Hospital Basel, University Basel, Basel, Switzerland
- Department of Neurosurgery, University Hospital Basel, University Basel, Basel, Switzerland
- Faculty of Medicine, University Basel, Basel, Switzerland
| |
Collapse
|
18
|
Liang M, Zhong H, Rong J, Li Y, Zhu C, Zhou L, Zhou R. Postnatal Lipopolysaccharide Exposure Impairs Adult Neurogenesis and Causes Depression-like Behaviors Through Astrocytes Activation Triggering GABAA Receptor Downregulation. Neuroscience 2019; 422:21-31. [DOI: 10.1016/j.neuroscience.2019.10.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 09/30/2019] [Accepted: 10/14/2019] [Indexed: 01/20/2023]
|
19
|
Abstract
Brain tissue lost after a stroke is not regenerated, although a repair response associated with neurogenesis does occur. A failure to regenerate functional brain tissue is not caused by the lack of available neural cells, but rather the absence of structural support to permit a repopulation of the lesion cavity. Inductive bioscaffolds can provide this support and promote the invasion of host cells into the tissue void. The putative mechanisms of bioscaffold degradation and its pivotal role to permit invasion of neural cells are reviewed and discussed in comparison to peripheral wound healing. Key differences between regenerating and non-regenerating tissues are contrasted in an evolutionary context, with a special focus on the neurogenic response as a conditio sine qua non for brain regeneration. The pivotal role of the immune system in biodegradation and the formation of a neovasculature are contextualized with regeneration of peripheral soft tissues. The application of rehabilitation to integrate newly forming brain tissue is suggested as necessary to develop functional tissue that can alleviate behavioral impairments. Pertinent aspects of brain tissue development are considered to provide guidance to produce a metabolically and functionally integrated de novo tissue. Although little is currently known about mechanisms involved in brain tissue regeneration, this review outlines the various components and their interplay to provide a framework for ongoing and future studies. It is envisaged that a better understanding of the mechanisms involved in brain tissue regeneration will improve the design of biomaterials and the methods used for implantation, as well as rehabilitation strategies that support the restoration of behavioral functions.
Collapse
Affiliation(s)
- Michel Modo
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States,Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, United States,Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States,Department of Radiology, University of Pittsburgh, Pittsburgh, PA, United States,*Correspondence: Michel Modo,
| |
Collapse
|
20
|
Functions of subventricular zone neural precursor cells in stroke recovery. Behav Brain Res 2019; 376:112209. [PMID: 31493429 DOI: 10.1016/j.bbr.2019.112209] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 08/11/2019] [Accepted: 09/03/2019] [Indexed: 12/16/2022]
Abstract
The proliferation and ectopic migration of neural precursor cells (NPCs) in response to ischemic brain injury was first reported two decades ago. Since then, studies of brain injury-induced subventricular zone cytogenesis, primarily in rodent models, have provided insight into the cellular and molecular determinants of this phenomenon and its modulation by various factors. However, despite considerable correlational evidence-and some direct evidence-to support contributions of NPCs to behavioral recovery after stroke, the causal mechanisms have not been identified. Here we discuss the subventricular zone cytogenic response and its possible roles in brain injury and disease, focusing on rodent models of stroke. Emerging evidence suggests that NPCs can modulate harmful responses and enhance reparative responses to neurologic diseases. We speculatively identify four broad functions of NPCs in the context of stroke: cell replacement, cytoprotection, remodeling of residual tissue, and immunomodulation. Thus, NPCs may have pleiotropic functions in supporting behavioral recovery after stroke.
Collapse
|
21
|
Genetic Risk for Rheumatoid Arthritis is Associated with Increased Striatal Volume in Healthy Young Adults. Sci Rep 2019; 9:10994. [PMID: 31358859 PMCID: PMC6662838 DOI: 10.1038/s41598-019-47505-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 06/24/2019] [Indexed: 01/11/2023] Open
Abstract
Rheumatoid arthritis (RA), an autoimmune disease, has recently been associated with increased striatal volume and decreased intracranial volume (ICV) in longstanding patients. As inflammation has been shown to precede the clinical diagnosis of RA and it is a known moderator of neuro- and gliogenesis, we were interested in testing whether these brain morphological changes appear before the clinical onset of disease in healthy young adult volunteers, as a function of relative genetic risk for RA. Genetic and structural MRI data were available for 516 healthy non-Hispanic Caucasian university students (275 women, mean age 19.78 ± 1.24 years). Polygenic risk scores were computed for each individual based on a genome-wide association study of RA, so that higher scores indicated higher risk. Striatal volume (sum of caudate, putamen, and nucleus accumbens volumes) and ICV were derived for each individual from high-resolution T1-weighted images. After controlling for sex, age, genetic components of ethnicity, socioeconomic status, and depressive symptoms, we found that higher RA polygenic risk scores were associated with increased striatal volume, but not decreased ICV. Our findings suggest that increased striatal volume may be linked to processes that precede disease onset, such as inflammation, while decreased ICV may relate to disease progression.
Collapse
|
22
|
The Anti-Inflammatory Effects of CXCR5 in the Mice Retina following Ischemia-Reperfusion Injury. BIOMED RESEARCH INTERNATIONAL 2019; 2019:3487607. [PMID: 31355256 PMCID: PMC6637708 DOI: 10.1155/2019/3487607] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 04/22/2019] [Accepted: 04/30/2019] [Indexed: 01/18/2023]
Abstract
Object Retinal ischemia-reperfusion (I/R) injury is a common pathological process in many ophthalmic diseases; there are no effective therapeutic approaches available currently. Increasing evidence indicates that microglia mediated neuroinflammation plays an important role in the retinal I/R injury. In this study, we aimed to investigate the roles of chemokine receptor CXCR5 in the pathological process of retinal I/R injury model. Method Retinal I/R injury model was established in CXCR5 knockout and wild mice by the acute elevation of intraocular pressure (AOH) for 60 minutes, and the eyes were harvested for further analyses. The cellular location of CXCR5 was detected by immunofluorescence staining; the expressions of CXCR5 and CXCL13 after I/R injury were analyzed by quantitative RT-PCR. The retinal microglia were detected as stained for Iba1 (+). Leakage of inflammatory cells was observed on the H&E stained cryosections. The protein expression and quantification of zonula occludens (ZO-1) were determined by Western blotting and densitometry. Capillary degeneration was identified on the intact retinal vasculatures prepared by trypsin digestion. Results The number of activated microglia marked by Iba1 antibody in the retina was increased after retinal I/R injury in both KO and WT mice, more significant in KO mice. The leakage of inflammatory cells was observed largely at 2 days after injury, but there was no or little leakage at 7 days. The number of inflammatory cells (mainly neutrophils) was greater in CXCR5 KO mice than in WT mice, mainly located under internal limiting membrane. CXCR5 deficiency led to more ZO-1 degradation in CXCR5 KO mice compared to C57BL6 WT mice 2 days after reperfusion. The cellular capillaries were also significantly increased in the KO mice compared to the WT mice. Conclusion Our findings suggest that the chemokine receptor CXCR5 may protect retina from ischemia-reperfusion injury by its anti-inflammatory effects. Thus, CXCR5 may be a promising therapeutic target for the treatment of retinal I/R injury.
Collapse
|
23
|
Covacu R, Brundin L. Endogenous spinal cord stem cells in multiple sclerosis and its animal model. J Neuroimmunol 2019; 331:4-10. [PMID: 27884460 DOI: 10.1016/j.jneuroim.2016.11.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 11/10/2016] [Accepted: 11/14/2016] [Indexed: 10/20/2022]
Abstract
The adult mammalian spinal cord (SC) harbors neural stem cells (NSCs). The SC-NSCs are mostly quiescent during physiological conditions but are quickly activated in traumatic injury models. The SC-NSCs generate mostly glia, but are able to differentiate into neurons when affected by favourable conditions. An example is the inflammatory milieu in the SC of rat EAE, where the SC-NSCs migrate into demyelinated lesions and give rise to both glia and neurons. In MS, cells with progenitor phenotypes accumulate in inflammatory lesions both in brain and SC, but the extent to which these cells contribute to repair remains to be revealed.
Collapse
Affiliation(s)
- Ruxandra Covacu
- Department of Clinical Neuroscience, Division of Neurology R3:04, Center of Molecular Medicine, L8:04, Karolinska Institutet, Stockholm, Sweden.
| | - Lou Brundin
- Department of Clinical Neuroscience, Division of Neurology R3:04, Center of Molecular Medicine, L8:04, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
24
|
Zhao L, Mulligan MK, Nowak TS. Substrain- and sex-dependent differences in stroke vulnerability in C57BL/6 mice. J Cereb Blood Flow Metab 2019; 39:426-438. [PMID: 29260927 PMCID: PMC6421252 DOI: 10.1177/0271678x17746174] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The C57BL/6 mouse strain is represented by distinct substrains, increasingly recognized to differ genetically and phenotypically. The current study compared stroke vulnerability among C57BL/6 J (J), C57BL/6JEiJ (JEiJ), C57BL/6ByJ (ByJ), C57BL/6NCrl (NCrl), C57BL/6NJ (NJ) and C57BL/6NTac (NTac) substrains, using a model of permanent distal middle cerebral artery and common carotid artery occlusion. Mean infarct volume was nearly two-fold smaller in J, JEiJ and ByJ substrains relative to NCrl, NJ and NTac (N-lineage) mice. This identifies a previously unrecognized confound in stroke studies involving genetically modified strain comparisons if control substrain background were not rigorously matched. Mean infarct size was smaller in females of J and ByJ substrains than in the corresponding males, but there was no sex difference for NCrl and NJ mice. A higher proportion of small infarcts in J and ByJ substrains was largely responsible for both substrain- and sex-dependent differences. These could not be straightforwardly explained by variations in posterior communicating artery patency, MCA anatomy or acute penumbral blood flow deficits. Their larger and more homogeneously distributed infarcts, together with their established use as the common background for many genetically modified strains, may make N-lineage C57BL/6 substrains the preferred choice for future studies in experimental stroke.
Collapse
Affiliation(s)
- Liang Zhao
- 1 Department of Neurology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Megan K Mulligan
- 2 Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Thaddeus S Nowak
- 1 Department of Neurology, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
25
|
Nowak TS, Mulligan MK. Impact of C57BL/6 substrain on sex-dependent differences in mouse stroke models. Neurochem Int 2018; 127:12-21. [PMID: 30448566 DOI: 10.1016/j.neuint.2018.11.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 11/14/2018] [Accepted: 11/14/2018] [Indexed: 01/18/2023]
Abstract
We have recently found significant variation in stroke vulnerability among substrains of C57BL/6 mice, observing that commonly used N-lineage substrains exhibit larger infarcts than C57BL/6J and related substrains. Parallel variation was also seen with respect to sex differences in stroke vulnerability, in that C57BL/6 mice of the N-lineage exhibited comparable infarct sizes in males and females, whereas infarcts tended to be smaller in females than in males of J-lineage substrains. This adds to the growing list of recognized phenotypic and genetic differences among C57BL/6 substrains. Although no previous studies have explicitly compared substrains with respect to sex differences in stroke vulnerability, unrecognized background mismatch has occurred in some studies involving control and genetically modified mice. The aims of this review are to: present the evidence for associated substrain- and sex-dependent differences in a mouse permanent occlusion stroke model; examine the extent to which the published literature in other models compares with these recent results; and consider the potential impact of unrecognized heterogeneity in substrain background on the interpretation of studies investigating the impact of genetic modifications on sex differences in stroke outcome. Substrain emerges as a critical variable to be documented in any experimental stroke study in mice.
Collapse
Affiliation(s)
- Thaddeus S Nowak
- Department of Neurology and Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA.
| | - Megan K Mulligan
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
26
|
Lin R, Lang M, Heinsinger N, Stricsek G, Zhang J, Iozzo R, Rosenwasser R, Iacovitti L. Stepwise impairment of neural stem cell proliferation and neurogenesis concomitant with disruption of blood-brain barrier in recurrent ischemic stroke. Neurobiol Dis 2018; 115:49-58. [PMID: 29605425 DOI: 10.1016/j.nbd.2018.03.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 03/12/2018] [Accepted: 03/28/2018] [Indexed: 01/15/2023] Open
Abstract
Stroke patients are at increased risk for recurrent stroke and development of post-stroke dementia. In this study, we investigated the effects of recurrent stroke on adult brain neurogenesis using a novel rat model of recurrent middle cerebral artery occlusion (MCAO) developed in our laboratory. Using BrdU incorporation, activation and depletion of stem cells in the subgranular zone (SGZ) and subventricular zone (SVZ) were assessed in control rats and rats after one or two strokes. In vitro neurosphere assay was used to assess the effects of plasma from normal and stroke rats. Also, EM and permeability studies were used to evaluate changes in the blood-brain-barrier (BBB) of the SGZ after recurrent stroke. We found that proliferation and neurogenesis was activated 14 days after MCAO. This was correlated with increased permeability in the BBB to factors which increase proliferation in a neurosphere assay. However, with each stroke, there was a stepwise decrease of proliferating stem cells and impaired neurogenesis on the ipsilateral side. On the contralateral side, this process stabilized after a first stroke. These studies indicate that stem cells are activated after MCAO, possibly after increased access to systemic stroke-related factors through a leaky BBB. However, the recruitment of stem cells for neurogenesis after stroke results in a stepwise ipsilateral decline with each ischemic event, which could contribute to post-stroke dementia.
Collapse
Affiliation(s)
- Ruihe Lin
- Department of Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA; The Joseph and Marie Field Cerebrovascular Research Laboratory, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA; Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Michael Lang
- Department of Neurological Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA; The Joseph and Marie Field Cerebrovascular Research Laboratory, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA; Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Nicolette Heinsinger
- Department of Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA; Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Geoffrey Stricsek
- Department of Neurological Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA; The Joseph and Marie Field Cerebrovascular Research Laboratory, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA; Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Justine Zhang
- Department of Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Renato Iozzo
- Department of Pathology, Anatomy, & Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Robert Rosenwasser
- Department of Neurological Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA; The Joseph and Marie Field Cerebrovascular Research Laboratory, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA; Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Lorraine Iacovitti
- Department of Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA; The Joseph and Marie Field Cerebrovascular Research Laboratory, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA; Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
27
|
Chen YA, Wang KC, Liu DZ, Young TH, Tsai LK. The Proliferation Capacity of Cultured Neural Stem Cells Promoted by CSF Collected from SAH Patients Correlates to Clinical Outcome. Sci Rep 2018; 8:1109. [PMID: 29348677 PMCID: PMC5773507 DOI: 10.1038/s41598-018-19371-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 12/29/2017] [Indexed: 11/29/2022] Open
Abstract
Neurogenesis from endogenous neural stem cells (NSCs) might contribute to functional recovery after stroke based on animal studies; however, the relationship between neurogenesis and post-stroke outcome has rarely been demonstrated in humans. We prospectively collected cerebrospinal fluid (CSF) from 36 patients with subarachnoid hemorrhage (SAH). The CSF was added to the culture medium of the rat NSCs to test the effects on proliferation (proliferation index [PI], percentage of Ki-67 immunoreactive cells). We correlated the PI with functional outcome based on the modified Rankin Scale at 3 months post-SAH. Treatment with the CSF samples collected from SAH patients showed a higher PI compared with those collected from patients with normal pressure hydrocephalus and untreated controls (20.3 ± 8.8 vs. 8.2 ± 5.1 and 7.8 ± 3.0, P < 0.001), indicating proliferation-promoting factors in CSF after SAH. The PI was positively correlated with SAH volume (p = 0.025). For patients with lower SAH volume, patients with favorable outcome had a higher PI than those with poor outcome (20.8 ± 6.9 vs. 14.6 ± 4.3, p = 0.047). Using multivariable logistic regression analysis, the PI was a positive determinant for favorable outcome (odds ratio, 1.17; 95% confidence interval, 1.00 to 1.36) that more proliferation-promoting factors in CSF was associated with better functional outcome in SAH patients.
Collapse
Affiliation(s)
- Yun-An Chen
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, Taiwan
| | - Kuo-Chuan Wang
- Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Der-Zen Liu
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Tai-Horng Young
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, Taiwan.
| | - Li-Kai Tsai
- Department of Neurology and Stroke Center, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan.
| |
Collapse
|
28
|
Song JJ, Oh SM, Kwon OC, Wulansari N, Lee HS, Chang MY, Lee E, Sun W, Lee SE, Chang S, An H, Lee CJ, Lee SH. Cografting astrocytes improves cell therapeutic outcomes in a Parkinson's disease model. J Clin Invest 2017; 128:463-482. [PMID: 29227284 DOI: 10.1172/jci93924] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 10/31/2017] [Indexed: 12/20/2022] Open
Abstract
Transplantation of neural progenitor cells (NPCs) is a potential therapy for treating neurodegenerative disorders, but this approach has faced many challenges and limited success, primarily because of inhospitable host brain environments that interfere with enriched neuron engraftment and function. Astrocytes play neurotrophic roles in the developing and adult brain, making them potential candidates for helping with modification of hostile brain environments. In this study, we examined whether astrocytic function could be utilized to overcome the current limitations of cell-based therapies in a murine model of Parkinson's disease (PD) that is characterized by dopamine (DA) neuron degeneration in the midbrain. We show here that cografting astrocytes, especially those derived from the midbrain, remarkably enhanced NPC-based cell therapeutic outcomes along with robust DA neuron engraftment in PD rats for at least 6 months after transplantation. We further show that engineering of donor astrocytes with Nurr1 and Foxa2, transcription factors that were recently reported to polarize harmful immunogenic glia into the neuroprotective form, further promoted the neurotrophic actions of grafted astrocytes in the cell therapeutic approach. Collectively, these findings suggest that cografting astrocytes could be a potential strategy for successful cell therapeutic outcomes in neurodegenerative disorders.
Collapse
Affiliation(s)
- Jae-Jin Song
- Department of Biochemistry and Molecular Biology, College of Medicine.,Hanyang Biomedical Research Institute, and.,Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea
| | - Sang-Min Oh
- Department of Biochemistry and Molecular Biology, College of Medicine.,Hanyang Biomedical Research Institute, and.,Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea
| | - Oh-Chan Kwon
- Department of Biochemistry and Molecular Biology, College of Medicine.,Hanyang Biomedical Research Institute, and.,Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea
| | - Noviana Wulansari
- Department of Biochemistry and Molecular Biology, College of Medicine.,Hanyang Biomedical Research Institute, and.,Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea
| | - Hyun-Seob Lee
- Genomic Core Facility, Transdisciplinary Research and Collaboration Division, Translational Research Institute, and.,Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea
| | - Mi-Yoon Chang
- Department of Biochemistry and Molecular Biology, College of Medicine.,Hanyang Biomedical Research Institute, and
| | - Eunsoo Lee
- Department of Anatomy and Division of Brain Korea 21 PLUS Program for Biomedical Science, Korea University College of Medicine, Seoul, South Korea
| | - Woong Sun
- Department of Anatomy and Division of Brain Korea 21 PLUS Program for Biomedical Science, Korea University College of Medicine, Seoul, South Korea
| | - Sang-Eun Lee
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
| | - Sunghoe Chang
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
| | - Heeyoung An
- Center for Neuroscience and.,Center for Glia-Neuron Interaction, Korea Institute of Science and Technology (KIST), Seoul, South Korea.,KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, South Korea
| | - C Justin Lee
- Center for Neuroscience and.,Center for Glia-Neuron Interaction, Korea Institute of Science and Technology (KIST), Seoul, South Korea
| | - Sang-Hun Lee
- Department of Biochemistry and Molecular Biology, College of Medicine.,Hanyang Biomedical Research Institute, and.,Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea
| |
Collapse
|
29
|
Wang J, Fu X, Zhang D, Yu L, Lu Z, Gao Y, Liu X, Man J, Li S, Li N, Wang M, Liu X, Chen X, Zang W, Yang Q, Wang J. Effects of crenolanib, a nonselective inhibitor of PDGFR, in a mouse model of transient middle cerebral artery occlusion. Neuroscience 2017; 364:202-211. [PMID: 28943249 PMCID: PMC5653447 DOI: 10.1016/j.neuroscience.2017.09.025] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 08/28/2017] [Accepted: 09/13/2017] [Indexed: 12/11/2022]
Abstract
Neurogenesis in the subventricular zone (SVZ) plays a vital role in neurologic recovery after stroke. However, only a small fraction of newly generated neuroblasts from the SVZ will survive long-term. Successful migration and survival of neuroblasts requires angiogenesis, lesion-derived chemo-attractants, and appropriate local microenvironments, which are partly regulated by the platelet-derived growth factor receptor (PDGFR) signaling pathway. In this study, we investigated the effects of PDGFR inhibition in a mouse model of transient middle cerebral artery occlusion (MCAO). We blocked the pathway using a nonselective PDGFR inhibitor, crenolanib, during the acute post-MCAO phase (days 1-3) or during the sub-acute phase (days 7-9). Downregulating the PDGFR signaling pathway with crenolanib from day 1 to day 3 after MCAO significantly decreased the migration of neuroblasts from the SVZ to the peri-infarct region, decreased angiogenesis, and lowered expression of vascular endothelial growth factor, stromal cell-derived factor-1, and monocyte chemotactic protein-1. Downregulation of the PDGFR signaling pathway on days 7-9 with crenolanib significantly increased apoptosis of the neuroblasts that had migrated to the peri-infarct region, increased the number of activated microglia, and decreased the expression of brain-derived neurotrophic factor, neurotrophin-3, and interleukin-10. Crenolanib treatment increased the apoptosis of pericytes and decreased the pericyte/vascular coverage, but had no effects on apoptosis of astrocytes. We conclude that the PDGFR signaling pathway plays a vital role in the SVZ neurogenesis after stroke. It can also affect angiogenesis, lesion-derived chemo-attractants, and the local microenvironment, which are all important to stroke-induced neurogenesis.
Collapse
Affiliation(s)
- Jianping Wang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Xiaojie Fu
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Di Zhang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lie Yu
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhengfang Lu
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yufeng Gao
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xianliang Liu
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jiang Man
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Sijia Li
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Nan Li
- Department of Neurology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Menghan Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xi Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xuemei Chen
- Department of Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Weidong Zang
- Department of Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Qingwu Yang
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Jian Wang
- Department of Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; Department of Anesthesiology/Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
30
|
MANF Promotes Differentiation and Migration of Neural Progenitor Cells with Potential Neural Regenerative Effects in Stroke. Mol Ther 2017; 26:238-255. [PMID: 29050872 PMCID: PMC5763030 DOI: 10.1016/j.ymthe.2017.09.019] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Revised: 08/31/2017] [Accepted: 09/18/2017] [Indexed: 01/05/2023] Open
Abstract
Cerebral ischemia activates endogenous reparative processes, such as increased proliferation of neural stem cells (NSCs) in the subventricular zone (SVZ) and migration of neural progenitor cells (NPCs) toward the ischemic area. However, this reparative process is limited because most of the NPCs die shortly after injury or are unable to arrive at the infarct boundary. In this study, we demonstrate for the first time that endogenous mesencephalic astrocyte-derived neurotrophic factor (MANF) protects NSCs against oxygen-glucose-deprivation-induced injury and has a crucial role in regulating NPC migration. In NSC cultures, MANF protein administration did not affect growth of cells but triggered neuronal and glial differentiation, followed by activation of STAT3. In SVZ explants, MANF overexpression facilitated cell migration and activated the STAT3 and ERK1/2 pathway. Using a rat model of cortical stroke, intracerebroventricular injections of MANF did not affect cell proliferation in the SVZ, but promoted migration of doublecortin (DCX)+ cells toward the corpus callosum and infarct boundary on day 14 post-stroke. Long-term infusion of MANF into the peri-infarct zone increased the recruitment of DCX+ cells in the infarct area. In conclusion, our data demonstrate a neuroregenerative activity of MANF that facilitates differentiation and migration of NPCs, thereby increasing recruitment of neuroblasts in stroke cortex.
Collapse
|
31
|
Falk S, Bugeon S, Ninkovic J, Pilz GA, Postiglione MP, Cremer H, Knoblich JA, Götz M. Time-Specific Effects of Spindle Positioning on Embryonic Progenitor Pool Composition and Adult Neural Stem Cell Seeding. Neuron 2017; 93:777-791.e3. [PMID: 28231465 PMCID: PMC5338691 DOI: 10.1016/j.neuron.2017.02.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 11/12/2016] [Accepted: 01/06/2017] [Indexed: 12/27/2022]
Abstract
The developmental mechanisms regulating the number of adult neural stem cells (aNSCs) are largely unknown. Here we show that the cleavage plane orientation in murine embryonic radial glia cells (RGCs) regulates the number of aNSCs in the lateral ganglionic eminence (LGE). Randomizing spindle orientation in RGCs by overexpression of Insc or a dominant-negative form of Lgn (dnLgn) reduces the frequency of self-renewing asymmetric divisions while favoring symmetric divisions generating two SNPs. Importantly, these changes during embryonic development result in reduced seeding of aNSCs. Interestingly, no effects on aNSC numbers were observed when Insc was overexpressed in postnatal RGCs or aNSCs. These data suggest a new mechanism for controlling aNSC numbers and show that the role of spindle orientation during brain development is highly time and region dependent. Randomization of the spindle orientation changes the progenitor pool composition Overexpression of Insc or dnLgn reduces asymmetric self-renewing division of aRGCs The change in embryonic progenitor pool leads to reduced seeding of adult NSCs Insc influences the seeding of adult NSCs in a narrow developmental time window
Collapse
Affiliation(s)
- Sven Falk
- Institute for Stem Cell Research, Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Neuherberg, Germany; Physiological Genomics, Biomedical Center, Ludwig-Maximilian University Munich, 82152 Planegg/Munich, Germany
| | - Stéphane Bugeon
- Aix-Marseille Université, Centre National de la Recherche Scientifique, IBDM, UMR7288, 13284 Marseille, France
| | - Jovica Ninkovic
- Institute for Stem Cell Research, Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Neuherberg, Germany; Physiological Genomics, Biomedical Center, Ludwig-Maximilian University Munich, 82152 Planegg/Munich, Germany
| | - Gregor-Alexander Pilz
- Institute for Stem Cell Research, Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Maria Pia Postiglione
- Institute of Molecular Biotechnology of the Austrian Academy of Science (IMBA), 1030 Vienna, Austria
| | - Harold Cremer
- Aix-Marseille Université, Centre National de la Recherche Scientifique, IBDM, UMR7288, 13284 Marseille, France
| | - Jürgen A Knoblich
- Institute of Molecular Biotechnology of the Austrian Academy of Science (IMBA), 1030 Vienna, Austria
| | - Magdalena Götz
- Institute for Stem Cell Research, Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Neuherberg, Germany; Physiological Genomics, Biomedical Center, Ludwig-Maximilian University Munich, 82152 Planegg/Munich, Germany; SYNERGY, Excellence Cluster of Systems Neurology, Biomedical Center, Ludwig-Maximilian University Munich, 82152 Planegg/Munich, Germany.
| |
Collapse
|
32
|
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: 22] [Impact Index Per Article: 2.8] [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
|
33
|
Zhang X, Zheng W, Wang T, Ren P, Wang F, Ma X, Wang J, Huang X. Danshen-Chuanxiong-Honghua Ameliorates Cerebral Impairment and Improves Spatial Cognitive Deficits after Transient Focal Ischemia and Identification of Active Compounds. Front Pharmacol 2017; 8:452. [PMID: 28769792 PMCID: PMC5513983 DOI: 10.3389/fphar.2017.00452] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 06/23/2017] [Indexed: 02/01/2023] Open
Abstract
Previously, we only apply a traditional Chinese medicine (TCM) Danshen-Chuanxiong-Honghua (DCH) for cardioprotection via anti-inflammation in rats of acute myocardial infarction by occluding coronary artery. Presently, we select not only DCH but also its main absorbed compound ferulic acid (FA) for cerebra protection via similar action of mechanism above in animals of the transient middle cerebral artery occlusion (tMCAO). We investigated whether oral administration of DCH and FA could ameliorate MCAO-induced brain lesions in animals. By using liquid chromatography-tandem mass spectrometry (LC-MS/MS), we analyzed four compounds, including tanshinol, salvianolic acid B, hydroxysafflor yellow A and especially FA as the putative active components of DCH extract in the plasma, cerebrospinal fluid and injured hippocampus of rats with MCAO. In our study, it was assumed that FA played a similar neuroprotective role to DCH. We found that oral pretreatment with DCH (10 or 20 g/kg) and FA (100 mg/kg) improved neurological function and alleviated the infarct volume as well as brain edema in a dose-dependent manner. These changes were accompanied by improved ischemia-induced apoptosis and decreased the inflammatory response. Additionally, chronic treatment with DCH reversed MCAO-induced spatial cognitive deficits in a manner associated with enhanced neurogenesis and increased the expression of brain-derived neurotrophic factor in lesions of the hippocampus. These findings suggest that DCH has the ability to recover cognitive impairment and offer neuroprotection against cerebral ischemic injury via inhibiting microenvironmental inflammation and triggering of neurogenesis in the hippocampus. FA could be one of the potential active compounds.
Collapse
Affiliation(s)
- Xianhua Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South UniversityChangsha, China.,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of PharmacogeneticsChangsha, China
| | - Wan Zheng
- Institute of TCM-Related Comorbid Depression, Nanjing University of Chinese MedicineNanjing, China
| | - Tingrui Wang
- Department of Neurology, Binzhou Central Hospital, Binzhou Medical CollegeBinzhou, China
| | - Ping Ren
- Institute of TCM-Related Comorbid Depression, Nanjing University of Chinese MedicineNanjing, China
| | - Fushun Wang
- School of Psychology, Nanjing University of Chinese MedicineNanjing, China
| | - Xinliang Ma
- Department of Emergency Medicine, Thomas Jefferson University, PhiladelphiaPA, United States
| | - Jian Wang
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, BaltimoreMD, United States
| | - Xi Huang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South UniversityChangsha, China.,Institute of TCM-Related Comorbid Depression, Nanjing University of Chinese MedicineNanjing, China
| |
Collapse
|
34
|
Jones KS, Connor B. Endogenous Brain Repair: Overriding intrinsic lineage determinates through injury-induced micro-environmental signals. NEUROGENESIS 2017; 4:1-5. [PMID: 28596976 DOI: 10.1080/23262133.2017.1297881] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 02/15/2017] [Accepted: 02/16/2017] [Indexed: 01/18/2023]
Abstract
Adult human neurogenesis has generated excitement over the last 2 decades with the idea that endogenous adult stem cells could act as a potential cell source for brain repair after injury. Indeed, many forms of experimentally induced brain injury including stroke and excitotoxic lesioning can promote proliferation from the subventricular zone and mobilise neuroblasts and oligodendrocyte progenitor cells to migrate through brain parenchyma to damaged regions. However the failure of neuroblasts to mature into appropriate neuronal subtypes for cell replacement has been an issue. Recent work by our group and others has indicated that micro-environmental signals released from areas of cell loss may be able to override intrinsic gene expression lineages and covert neuroblasts into oligodendrocyte progenitor cells. This commentary will discuss the enhanced fate plasticity of both adult neural progenitors and parenchymal NG2 cells after injury, and the importance of understanding brain-injury induced micro-environmental signals in the quest toward promoting endogenous regeneration after injury.
Collapse
Affiliation(s)
- Kathryn S Jones
- Department of Pharmacology and Clinical Pharmacology, Centre for Brain Research, School of Medical Sciences, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Bronwen Connor
- Department of Pharmacology and Clinical Pharmacology, Centre for Brain Research, School of Medical Sciences, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
35
|
Tajiri N, Quach DM, Kaneko Y, Wu S, Lee D, Lam T, Hayama KL, Hazel TG, Johe K, Wu MC, Borlongan CV. NSI-189, a small molecule with neurogenic properties, exerts behavioral, and neurostructural benefits in stroke rats. J Cell Physiol 2017; 232:2731-2740. [PMID: 28181668 PMCID: PMC5518191 DOI: 10.1002/jcp.25847] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 02/07/2017] [Indexed: 12/26/2022]
Abstract
Enhancing neurogenesis may be a powerful stroke therapy. Here, we tested in a rat model of ischemic stroke the beneficial effects of NSI-189, an orally active, new molecular entity (mol. wt. 366) with enhanced neurogenic activity, and indicated as an anti-depressant drug in a clinical trial (Fava et al., , Molecular Psychiatry, DOI: 10.1038/mp.2015.178) and being tested in a Phase 2 efficacy trial (ClinicalTrials.gov, , ClinicalTrials.gov Identifier: NCT02695472) for treatment of major depression. Oral administration of NSI-189 in adult Sprague-Dawley rats starting at 6 hr after middle cerebral artery occlusion, and daily thereafter over the next 12 weeks resulted in significant amelioration of stroke-induced motor and neurological deficits, which was maintained up to 24 weeks post-stroke. Histopathological assessment of stroke brains from NSI-189-treated animals revealed significant increments in neurite outgrowth as evidenced by MAP2 immunoreactivity that was prominently detected in the hippocampus and partially in the cortex. These results suggest NSI-189 actively stimulated remodeling of the stroke brain. Parallel in vitro studies further probed this remodeling process and demonstrated that oxygen glucose deprivation and reperfusion (OGD/R) initiated typical cell death processes, which were reversed by NSI-189 treatment characterized by significant attenuation of OGD/R-mediated hippocampal cell death and increased Ki67 and MAP2 expression, coupled with upregulation of neurogenic factors such as BDNF and SCF. These findings support the use of oral NSI-189 as a therapeutic agent well beyond the initial 6-hr time window to accelerate and enhance the overall functional improvement in the initial 6 months post stroke.
Collapse
Affiliation(s)
- Naoki Tajiri
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University South Florida College of Medicine, Tampa, Florida
| | | | - Yuji Kaneko
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University South Florida College of Medicine, Tampa, Florida
| | | | - David Lee
- Neuralstem, Inc., Rockville, Maryland
| | - Tina Lam
- Neuralstem, Inc., Rockville, Maryland
| | | | | | - Karl Johe
- Neuralstem, Inc., Rockville, Maryland
| | | | - Cesar V Borlongan
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University South Florida College of Medicine, Tampa, Florida
| |
Collapse
|
36
|
Hay M, Vanderah TW, Samareh-Jahani F, Constantopoulos E, Uprety AR, Barnes CA, Konhilas J. Cognitive impairment in heart failure: A protective role for angiotensin-(1-7). Behav Neurosci 2017; 131:99-114. [PMID: 28054808 DOI: 10.1037/bne0000182] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Patients with congestive heart failure (CHF) have increased hospital readmission rates and mortality if they are concomitantly diagnosed with cognitive decline and memory loss. Accordingly, we developed a preclinical model of CHF-induced cognitive impairment with the goal of developing novel protective therapies against CHF related cognitive decline. CHF was induced by ligation of the left coronary artery to instigate a myocardial infarction (MI). By 4- and 8-weeks post-MI, CHF mice had approximately a 50% and 70% decline in ejection fraction as measured by echocardiography. At both 4- and 8-weeks post-MI, spatial memory performance in CHF mice as tested using the Morris water task was significantly impaired as compared with sham. In addition, CHF mice had significantly worse performance on object recognition when compared with shams as measured by discrimination ratios during the novel object recognition NOR task. At 8-weeks post-MI, a subgroup of CHF mice were given Angiotensin (Ang)-(1-7) (50mcg/kg/hr) subcutaneously for 4 weeks. Following 3 weeks treatment with systemic Ang-(1-7), the CHF mice NOR discrimination ratios were similar to shams and significantly better than the performance of CHF mice treated with saline. Ang-(1-7) also improved spatial memory in CHF mice as compared with shams. Ang-(1-7) had no effect on cardiac function. Inflammatory biomarker studies from plasma revealed a pattern of neuroprotection that may underlie the observed improvements in cognition. These results demonstrate a preclinical mouse model of CHF that exhibits both spatial memory and object recognition dysfunction. Furthermore, this CHF-induced cognitive impairment is attenuated by treatment with systemic Ang-(1-7). (PsycINFO Database Record
Collapse
Affiliation(s)
| | | | | | | | - Ajay R Uprety
- Evelyn F. McKnight Brain Institute, University of Arizona
| | - Carol A Barnes
- Evelyn F. McKnight Brain Institute, University of Arizona
| | - John Konhilas
- Department of Physiology and Sarver Heart Center, University of Arizona
| |
Collapse
|
37
|
Llorens-Bobadilla E, Martin-Villalba A. Adult NSC diversity and plasticity: the role of the niche. Curr Opin Neurobiol 2016; 42:68-74. [PMID: 27978480 DOI: 10.1016/j.conb.2016.11.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 11/22/2016] [Accepted: 11/22/2016] [Indexed: 12/20/2022]
Abstract
Adult somatic stem cells are generally defined as cells with the ability to differentiate into multiple different lineages and to self-renew during long periods of time. These features were long presumed to be represented in one single tissue-specific stem cell. Recent development of single-cell technologies reveals the existence of diversity in fate and activation state of somatic stem cells within the blood, skin and intestinal compartments [1] but also in the adult brain. Here we review how recent advances have expanded our view of neural stem cells (NSCs) as a diverse pool of cells and how the specialized microenvironment in which they reside acts to maintain this diversity. In addition, we discuss the plasticity of the system in the injured brain.
Collapse
Affiliation(s)
- Enric Llorens-Bobadilla
- Department of Molecular Neurobiology, German Cancer Research Center (DFKZ), Heidelberg, Germany.
| | - Ana Martin-Villalba
- Department of Molecular Neurobiology, German Cancer Research Center (DFKZ), Heidelberg, Germany.
| |
Collapse
|
38
|
Okuneva O, Li Z, Körber I, Tegelberg S, Joensuu T, Tian L, Lehesjoki AE. Brain inflammation is accompanied by peripheral inflammation in Cstb -/- mice, a model for progressive myoclonus epilepsy. J Neuroinflammation 2016; 13:298. [PMID: 27894304 PMCID: PMC5127053 DOI: 10.1186/s12974-016-0764-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 11/16/2016] [Indexed: 01/16/2023] Open
Abstract
Progressive myoclonus epilepsy of Unverricht-Lundborg type (EPM1) is an autosomal recessively inherited childhood-onset neurodegenerative disorder, characterized by myoclonus, seizures, and ataxia. Mutations in the cystatin B gene (CSTB) underlie EPM1. The CSTB-deficient (Cstb -/- ) mouse model recapitulates key features of EPM1, including myoclonic seizures. The mice show early microglial activation that precedes seizure onset and neuronal loss and leads to neuroinflammation. We here characterized the inflammatory phenotype of Cstb -/- mice in more detail. We found higher concentrations of chemokines and pro-inflammatory cytokines in the serum of Cstb -/- mice and higher CXCL13 expression in activated microglia in Cstb -/- compared to control mouse brains. The elevated chemokine levels were not accompanied by blood-brain barrier disruption, despite increased brain vascularization. Macrophages in the spleen and brain of Cstb -/- mice were predominantly pro-inflammatory. Taken together, these data show that CXCL13 expression is a hallmark of microglial activation in Cstb -/- mice and that the brain inflammation is linked to peripheral inflammatory changes, which might contribute to the disease pathology of EPM1.
Collapse
Affiliation(s)
- Olesya Okuneva
- Folkhälsan Institute of Genetics, Haartmaninkatu 8, 00014 Helsinki, Finland
- Research Program’s Unit, Molecular Neurology, University of Helsinki, Haartmaninkatu 8, 00014 Helsinki, Finland
- Neuroscience Center, University of Helsinki, Viikinkaari 4, 00014 Helsinki, Finland
| | - Zhilin Li
- Neuroscience Center, University of Helsinki, Viikinkaari 4, 00014 Helsinki, Finland
| | - Inken Körber
- Folkhälsan Institute of Genetics, Haartmaninkatu 8, 00014 Helsinki, Finland
- Research Program’s Unit, Molecular Neurology, University of Helsinki, Haartmaninkatu 8, 00014 Helsinki, Finland
- Neuroscience Center, University of Helsinki, Viikinkaari 4, 00014 Helsinki, Finland
| | - Saara Tegelberg
- Folkhälsan Institute of Genetics, Haartmaninkatu 8, 00014 Helsinki, Finland
- Research Program’s Unit, Molecular Neurology, University of Helsinki, Haartmaninkatu 8, 00014 Helsinki, Finland
- Neuroscience Center, University of Helsinki, Viikinkaari 4, 00014 Helsinki, Finland
| | - Tarja Joensuu
- Folkhälsan Institute of Genetics, Haartmaninkatu 8, 00014 Helsinki, Finland
- Research Program’s Unit, Molecular Neurology, University of Helsinki, Haartmaninkatu 8, 00014 Helsinki, Finland
- Neuroscience Center, University of Helsinki, Viikinkaari 4, 00014 Helsinki, Finland
| | - Li Tian
- Neuroscience Center, University of Helsinki, Viikinkaari 4, 00014 Helsinki, Finland
- Beijing Huilongguan Hospital, Peking University teaching hospital, Beijing, China
| | - Anna-Elina Lehesjoki
- Folkhälsan Institute of Genetics, Haartmaninkatu 8, 00014 Helsinki, Finland
- Research Program’s Unit, Molecular Neurology, University of Helsinki, Haartmaninkatu 8, 00014 Helsinki, Finland
- Neuroscience Center, University of Helsinki, Viikinkaari 4, 00014 Helsinki, Finland
| |
Collapse
|
39
|
Early Postnatal Lipopolysaccharide Exposure Leads to Enhanced Neurogenesis and Impaired Communicative Functions in Rats. PLoS One 2016; 11:e0164403. [PMID: 27723799 PMCID: PMC5056722 DOI: 10.1371/journal.pone.0164403] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 09/23/2016] [Indexed: 11/23/2022] Open
Abstract
Perinatal infection is a well-identified risk factor for a number of neurodevelopmental disorders, including brain white matter injury (WMI) and Autism Spectrum Disorders (ASD). The underlying mechanisms by which early life inflammatory events cause aberrant neural, cytoarchitectural, and network organization, remain elusive. This study is aimed to investigate how systemic lipopolysaccharide (LPS)-induced neuroinflammation affects microglia phenotypes and early neural developmental events in rats. We show here that LPS exposure at early postnatal day 3 leads to a robust microglia activation which is characterized with mixed microglial proinflammatory (M1) and anti-inflammatory (M2) phenotypes. More specifically, we found that microglial M1 markers iNOS and MHC-II were induced at relatively low levels in a regionally restricted manner, whereas M2 markers CD206 and TGFβ were strongly upregulated in a sub-set of activated microglia in multiple white and gray matter structures. This unique microglial response was associated with a marked decrease in naturally occurring apoptosis, but an increase in cell proliferation in the subventricular zone (SVZ) and the dentate gyrus (DG) of hippocampus. LPS exposure also leads to a significant increase in oligodendrocyte lineage population without causing discernible hypermyelination. Moreover, LPS-exposed rats exhibited significant impairments in communicative and cognitive functions. These findings suggest a possible role of M2-like microglial activation in abnormal neural development that may underlie ASD-like behavioral impairments.
Collapse
|
40
|
Abstract
Neural stem/progenitor cells (NSCs/NPCs) are present in different locations in the central nervous system. In the subgranular zone (SGZ) there is a constant generation of new neurons under normal conditions. New neurons are also formed from the subventricular zone (SVZ) NSCs, and they migrate anteriorly as neuroblast to the olfactory bulb in rodents, whereas in humans migration is directed toward striatum. Most CNS injuries elicit proliferation and migration of the NSCs toward the injury site, indicating the activation of a regenerative response. However, regeneration from NSC is incomplete, and this could be due to detrimental cues encountered during inflammation. Different CNS diseases and trauma cause activation of the innate and adaptive immune responses that influence the NSCs. Furthermore, NSCs in the brain react differently to inflammatory cues than their counterparts in the spinal cord. In this review, we have summarized the effects of inflammation on NSCs in relation to their origin and briefly described the NSC activity during different neurological diseases or experimental models.
Collapse
Affiliation(s)
- Ruxandra Covacu
- 1 Depatment of Clinical Neuroscience, Neurology, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| | - Lou Brundin
- 1 Depatment of Clinical Neuroscience, Neurology, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
41
|
Xiong XY, Liu L, Yang QW. Functions and mechanisms of microglia/macrophages in neuroinflammation and neurogenesis after stroke. Prog Neurobiol 2016; 142:23-44. [PMID: 27166859 DOI: 10.1016/j.pneurobio.2016.05.001] [Citation(s) in RCA: 499] [Impact Index Per Article: 55.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 04/20/2016] [Accepted: 05/01/2016] [Indexed: 02/08/2023]
Abstract
Microglia/macrophages are the major immune cells involved in the defence against brain damage. Their morphology and functional changes are correlated with the release of danger signals induced by stroke. These cells are normally responsible for clearing away dead neural cells and restoring neuronal functions. However, when excessively activated by the damage-associated molecular patterns following stroke, they can produce a large number of proinflammatory cytokines that can disrupt neural cells and the blood-brain barrier and influence neurogenesis. These effects indicate the important roles of microglia/macrophages in the pathophysiological processes of stroke. However, the modifiable and adaptable nature of microglia/macrophages may also be beneficial for brain repair and not just result in damage. These distinct roles may be attributed to the different microglia/macrophage phenotypes because the M1 population is mainly destructive, while the M2 population is neuroprotective. Additionally, different gene expression signature changes in microglia/macrophages have been found in diverse inflammatory milieus. These biofunctional features enable dual roles for microglia/macrophages in brain damage and repair. Currently, it is thought that the proper inflammatory milieu may provide a suitable microenvironment for neurogenesis; however, detailed mechanisms underlying the inflammatory responses that initiate or inhibit neurogenesis remain unknown. This review summarizes recent progress concerning the mechanisms involved in brain damage, repair and regeneration related to microglia/macrophage activation and phenotype transition after stroke. We also argue that future translational studies should be targeting multiple key regulating molecules to improve brain repair, which should be accompanied by the concept of a "therapeutic time window" for sequential therapies.
Collapse
Affiliation(s)
- Xiao-Yi Xiong
- Department of Neurology, Xinqiao Hospital & The Second Affiliated Hospital, The Third Military Medical University, Xinqiao zhengjie No.183, Shapingba District Chongqing, 400037, China
| | - Liang Liu
- Department of Neurology, Xinqiao Hospital & The Second Affiliated Hospital, The Third Military Medical University, Xinqiao zhengjie No.183, Shapingba District Chongqing, 400037, China
| | - Qing-Wu Yang
- Department of Neurology, Xinqiao Hospital & The Second Affiliated Hospital, The Third Military Medical University, Xinqiao zhengjie No.183, Shapingba District Chongqing, 400037, China.
| |
Collapse
|
42
|
Pérez-Martín M, Rivera P, Blanco E, Lorefice C, Decara J, Pavón FJ, Serrano A, Rodríguez de Fonseca F, Suárez J. Environmental Enrichment, Age, and PPARα Interact to Regulate Proliferation in Neurogenic Niches. Front Neurosci 2016; 10:89. [PMID: 27013951 PMCID: PMC4783391 DOI: 10.3389/fnins.2016.00089] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 02/22/2016] [Indexed: 11/13/2022] Open
Abstract
Peroxisome proliferator-activated receptor alpha (PPARα) ligands have been shown to modulate recovery after brain insults such as ischemia and irradiation by enhancing neurogenesis. In the present study, we investigated the effect of the genetic deletion of PPARα receptors on the proliferative rate of neural precursor cells (NPC) in the adult brain. The study was performed in aged Pparα−/− mice exposed to nutritional (treats) and environmental (games) enrichments for 20 days. We performed immunohistochemical analyses of cells containing the replicating cell DNA marker 5-bromo-2′-deoxyuridine (BrdU+) and the immature neuronal marker doublecortin (Dcx+) in the main neurogenic zones of the adult brain: subgranular zone of dentate gyrus (SGZ), subventricular zone of lateral ventricles (SVZ), and/or hypothalamus. Results indicated a reduction in the number of BrdU+ cells in the neurogenic zones analyzed as well as Dcx+ cells in the SGZ during aging (2, 6, and 18 months). Pparα deficiency alleviated the age-related reduction of NPC proliferation (BrdU+ cells) in the SVZ of the 18-months-old mice. While no genotype effect on NPC proliferation was detected in the SGZ during aging, an accentuated reduction in the number of Dcx+ cells was observed in the SGZ of the 6-months-old Pparα−/− mice. Exposing the 18-months-old mice to nutritional and environmental enrichments reversed the Pparα−/−-induced impairment of NPC proliferation in the neurogenic zones analyzed. The enriched environment did not modify the number of SGZ Dcx+ cells in the 18 months old Pparα−/− mice. These results identify PPARα receptors as a potential target to counteract the naturally observed decline in adult NPC proliferation associated with aging and impoverished environments.
Collapse
Affiliation(s)
- Margarita Pérez-Martín
- Departamento de Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga Málaga, Spain
| | - Patricia Rivera
- UGC Salud Mental, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga-Hospital Universitario Regional de Málaga Málaga, Spain
| | - Eduardo Blanco
- UGC Salud Mental, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga-Hospital Universitario Regional de MálagaMálaga, Spain; Departament de Pedagogia i Psicologia, Facultat d'Educació, Psicologia i Treball Social, Universitat de LleidaLleida, Spain
| | - Clara Lorefice
- Departamento de Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga, Universidad de MálagaMálaga, Spain; UGC Salud Mental, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga-Hospital Universitario Regional de MálagaMálaga, Spain
| | - Juan Decara
- UGC Salud Mental, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga-Hospital Universitario Regional de Málaga Málaga, Spain
| | - Francisco J Pavón
- UGC Salud Mental, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga-Hospital Universitario Regional de Málaga Málaga, Spain
| | - Antonia Serrano
- UGC Salud Mental, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga-Hospital Universitario Regional de Málaga Málaga, Spain
| | - Fernando Rodríguez de Fonseca
- UGC Salud Mental, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga-Hospital Universitario Regional de Málaga Málaga, Spain
| | - Juan Suárez
- UGC Salud Mental, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga-Hospital Universitario Regional de Málaga Málaga, Spain
| |
Collapse
|
43
|
Amantea D, Bagetta G. Drug repurposing for immune modulation in acute ischemic stroke. Curr Opin Pharmacol 2016; 26:124-30. [DOI: 10.1016/j.coph.2015.11.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 11/11/2015] [Accepted: 11/16/2015] [Indexed: 12/24/2022]
|