1
|
Ren X, He J, Hu H, Kohsaka S, Zhao LR. Hematopoietic growth factors Regulate Entry of Monocytes into the Adult Brain via Chemokine Receptor CCR5. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.15.594359. [PMID: 38798506 PMCID: PMC11118552 DOI: 10.1101/2024.05.15.594359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Monocytes are circulating macrophage precursors and are generated from bone marrow hematopoietic stem cells. In the adults, monocytes continuously replenish cerebral border-associated macrophages under a physiological condition. Monocytes also rapidly infiltrate into the brain in the settings of pathological conditions. The mechanisms of recruiting monocyte-derived macrophages into the brain under pathological conditions have been extensively studied. However, it remains unclear how monocytes enter the brain for renewal of border-associated macrophages under the physiological condition. Using both in vitro and in vivo approaches, this study reveals that the combination of two hematopoietic growth factors, stem cell factor (SCF) and granulocyte colony-stimulating factor (G-CSF), complementarily and synergistically enhances adhesion of monocytes to cerebral endothelial cells in a dose dependent manner. Cysteine-cysteine chemokine receptor 5 (CCR5) in brain endothelial cells, but not cell adhesion molecules mediating neuroinflammation-related infiltration of monocyte-derived macrophages, modulates the SCF+G-CSF-enhanced monocyte-endothelial cell adhesion. Blocking CCR5 or genetically deleting CCR5 reduces monocyte-endothelial cell adhesion induced by SCF+G-CSF. SCF+G-CSF-enhanced recruitment of bone marrow-derived monocytes/macrophages in cerebral perivascular space is also reduced in adult CCR5 knockout mice. This study demonstrates the contribution of SCF and G-CSF in regulating the entry of monocytes into the adult brain to replenish perivascular macrophages.
Collapse
|
2
|
Chang J, Li Y, Shan X, Chen X, Yan X, Liu J, Zhao L. Neural stem cells promote neuroplasticity: a promising therapeutic strategy for the treatment of Alzheimer's disease. Neural Regen Res 2024; 19:619-628. [PMID: 37721293 PMCID: PMC10581561 DOI: 10.4103/1673-5374.380874] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 04/04/2023] [Accepted: 06/10/2023] [Indexed: 09/19/2023] Open
Abstract
Recent studies have demonstrated that neuroplasticity, such as synaptic plasticity and neurogenesis, exists throughout the normal lifespan but declines with age and is significantly impaired in individuals with Alzheimer's disease. Hence, promoting neuroplasticity may represent an effective strategy with which Alzheimer's disease can be alleviated. Due to their significant ability to self-renew, differentiate, and migrate, neural stem cells play an essential role in reversing synaptic and neuronal damage, reducing the pathology of Alzheimer's disease, including amyloid-β, tau protein, and neuroinflammation, and secreting neurotrophic factors and growth factors that are related to plasticity. These events can promote synaptic plasticity and neurogenesis to repair the microenvironment of the mammalian brain. Consequently, neural stem cells are considered to represent a potential regenerative therapy with which to improve Alzheimer's disease and other neurodegenerative diseases. In this review, we discuss how neural stem cells regulate neuroplasticity and optimize their effects to enhance their potential for treating Alzheimer's disease in the clinic.
Collapse
Affiliation(s)
- Jun Chang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yujiao Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Xiaoqian Shan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Xi Chen
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Xuhe Yan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Jianwei Liu
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lan Zhao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| |
Collapse
|
3
|
Bazarnyi VV, Kovtun OP, Koryakina OV, Polushina LG, Maksimova AY. [A pilot study of cytokine profile in cerebrospinal fluid of children with acute lymphocytic leukemia and neurotoxic side effects of chemotherapy]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2021; 67:374-377. [PMID: 34414897 DOI: 10.18097/pbmc20216704374] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
In some cases standard chemotherapy of acute lymphocytic leukemia (ALL) leads to neurotoxicity; its mechanisms, methods of prognosis, and prevention are being actively studied. The aim of this study was to assess the cytokine profile in cerebrospinal fluid (CSF) of children with ALL and neurotoxic side effects of chemotherapy. This prospective study included 24 children with ALL aged from 3 to 17 years. Patients were further subdivided into ALL patients with (main group) and without neurological complications (comparison group). The level of cytokines in CSF was measured by Xmap technology (Luminex) using Invitrogen test systems (eBioscience) and the Luminex 200 system. The comparative analysis of the cytokine profile in the group of children with chemotherapy-induced neurotoxic complications revealed elevated levels of chemokine CXCL12 (SDF-1α) and stem cell factor (SCF). Increased level of these cytokines in CSF was characterized by a relatively risk for development of toxic peripheral neuropathy.
Collapse
Affiliation(s)
- V V Bazarnyi
- Ural State Medical University, Yekaterinburg, Russia
| | - O P Kovtun
- Ural State Medical University, Yekaterinburg, Russia
| | - O V Koryakina
- Ural State Medical University, Yekaterinburg, Russia; Regional Children's Clinical Hospital, Yekaterinburg, Russia
| | - L G Polushina
- Ural State Medical University, Yekaterinburg, Russia
| | | |
Collapse
|
4
|
Pang CY, Yang KL, Fu CH, Sun LY, Chen SY, Liao CH. G-CSF enhances the therapeutic potency of stem cells transplantation in spinal cord-injured rats. Regen Med 2019; 14:571-583. [DOI: 10.2217/rme-2018-0104] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Aim: The therapeutic effects of human wisdom teeth-derived neuronal stem cell (tNSC) cotreatment with granulocyte-colony-stimulating factor (G-CSF) were evaluated for contusion-induced spinal cord injury in rats. Materials & methods: 7 days after contusion, tNSCs were transplanted to the injury site and followed by G-CSF cotreatment for 5 days. Behavioral deficits were evaluated by the Basso, Beattie and Bresnahan test. The injury site was collected for immunohistochemistry analysis. Results: The Basso, Beattie and Bresnahan test significantly improved in the cotreated group compared with the tNSCs or G-CSF single treatment groups. However, inflammation indices did not differ among the three groups. In vitro experiment demonstrated that tNSCs express both G-CSF and its relevant receptor. G-CSF enhanced tNSC proliferation and neurotrophins secretion in vitro. Conclusion: This study demonstrated that G-CSF enhances neurotrophins secretion of tNSCs, and might help improving functional recovery from spinal cord injury in rats if they were given together.
Collapse
Affiliation(s)
- Cheng-Yoong Pang
- Department of Medical Research, Hualien Tzu Chi Hospital, Hualien, Taiwan 970
- Cardiovascular & Metabolomics Research Center, Hualien Tzu Chi Hospital, Hualien, Taiwan 970
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan 970
| | - Kuo-Liang Yang
- Buddhist Tzu Chi Stem Cells Center, Hualien Tzu Chi Hospital, Hualien, Taiwan 970
- Tzu Chi Cord Blood Bank, Hualien Tzu Chi Hospital, Hualien, Taiwan 970
| | - Chin-Hua Fu
- Department of Neurology, Taichung Tzu Chi Hospital, Taichung, Taiwan 427
| | - Li-Yi Sun
- Department of Medical Research, Hualien Tzu Chi Hospital, Hualien, Taiwan 970
- Gene & Stem Cell Production Center, Hualien Tzu Chi Hospital, Hualien, Taiwan 970
| | - Shin-Yuan Chen
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Hualien, Taiwan 970
| | - Chia-Hsin Liao
- Department of Medical Research, Hualien Tzu Chi Hospital, Hualien, Taiwan 970
- Department of Nature Science, Holistic Education Center, Tzu Chi University of Science & Technology, Hualien, Taiwan 970
| |
Collapse
|
5
|
Zhao LR, Willing A. Enhancing endogenous capacity to repair a stroke-damaged brain: An evolving field for stroke research. Prog Neurobiol 2018; 163-164:5-26. [PMID: 29476785 PMCID: PMC6075953 DOI: 10.1016/j.pneurobio.2018.01.004] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 01/11/2018] [Accepted: 01/30/2018] [Indexed: 02/07/2023]
Abstract
Stroke represents a severe medical condition that causes stroke survivors to suffer from long-term and even lifelong disability. Over the past several decades, a vast majority of stroke research targets neuroprotection in the acute phase, while little work has been done to enhance stroke recovery at the later stage. Through reviewing current understanding of brain plasticity, stroke pathology, and emerging preclinical and clinical restorative approaches, this review aims to provide new insights to advance the research field for stroke recovery. Lifelong brain plasticity offers the long-lasting possibility to repair a stroke-damaged brain. Stroke impairs the structural and functional integrity of entire brain networks; the restorative approaches containing multi-components have great potential to maximize stroke recovery by rebuilding and normalizing the stroke-disrupted entire brain networks and brain functioning. The restorative window for stroke recovery is much longer than previously thought. The optimal time for brain repair appears to be at later stage of stroke rather than the earlier stage. It is expected that these new insights will advance our understanding of stroke recovery and assist in developing the next generation of restorative approaches for enhancing brain repair after stroke.
Collapse
Affiliation(s)
- Li-Ru Zhao
- Department of Neurosurgery, State University of New York, Upstate Medical University, Syracuse, NY, 13210, USA.
| | - Alison Willing
- Center for Excellence in Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL, 33612, USA.
| |
Collapse
|
6
|
Turning Death to Growth: Hematopoietic Growth Factors Promote Neurite Outgrowth through MEK/ERK/p53 Pathway. Mol Neurobiol 2017; 55:5913-5925. [PMID: 29119536 DOI: 10.1007/s12035-017-0814-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 10/27/2017] [Indexed: 12/23/2022]
Abstract
Stem cell factor (SCF) and granulocyte colony-stimulating factor (G-CSF) are the essential hematopoietic growth factors to control hematopoiesis. However, the role of SCF and G-CSF in the central nervous system remains poorly understood. Here, we have demonstrated the involvement of MEK/ERK/p53 signaling in SCF + G-CSF-enhanced neurite extension. Cortical neurons dissected from embryonic rat brains were seeded onto the membranes of transwell inserts, and neurite outgrowth was determined by using both the neurite outgrowth quantification assay kit and immunostaining of β III tubulin. Quantitative RT-PCR and western blotting were used for determining gene and protein expression of ERK and p53, respectively. p53 small interfering RNA (siRNAs) were introduced into neurons for examining the involvement of p53 in SCF + G-CSF-mediated neurite outgrowth. We observed that both SCF and G-CSF alone increased activation of MEK/ERK and gene expression of p53, while SCF + G-CSF synergistically activated the MEK/ERK signaling and upregulated p53 expression. MEK specific inhibitors (PD98059 and U0126) blocked the SCF + G-CSF-increased ERK phosphorylation and p53 gene and protein expression, and the MEK specific inhibitors also eliminated the SCF + G-CSF-promoted neurite outgrowth. p53 siRNAs knocked down the SCF + G-CSF-elevated p53 protein and prevented the SCF + G-CSF-enhanced neurite outgrowth. These findings suggest that activation of MEK/ERK/p53 signaling is required for SCF + G-CSF-promoted neurite outgrowth. Through the pro-apoptotic pathway of the MEK/ERK/p53, SCF + G-CSF turns neuronal fate from apoptotic commitment toward neural network generation. This observation provides novel insights into the putative role of SCF + G-CSF in supporting generation of neural connectivity during CNS development and in brain repair under pathological or neurodegenerative conditions.
Collapse
|
7
|
Liska MG, Dela Peña I. Granulocyte-colony stimulating factor and umbilical cord blood cell transplantation: Synergistic therapies for the treatment of traumatic brain injury. Brain Circ 2017; 3:143-151. [PMID: 30276316 PMCID: PMC6057694 DOI: 10.4103/bc.bc_19_17] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Revised: 08/31/2017] [Accepted: 09/05/2017] [Indexed: 02/06/2023] Open
Abstract
Traumatic brain injury (TBI) is now characterized as a progressive, degenerative disease and continues to stand as a prevalent cause of death and disability. The pathophysiology of TBI is complex, with a variety of secondary cell death pathways occurring which may persist chronically following the initial cerebral insult. Current therapeutic options for TBI are minimal, with surgical intervention or rehabilitation therapy existing as the only viable treatments. Considering the success of stem-cell therapies in various other neurological diseases, their use has been proposed as a potential potent therapy for patients suffering TBI. Moreover, stem cells are highly amenable to adjunctive use with other therapies, providing an opportunity to overcome the inherent limitations of using a single therapeutic agent. Our research has verified this additive potential by demonstrating the efficacy of co-delivering human umbilical cord blood (hUCB) cells with granulocyte-colony stimulating factor (G-CSF) in a murine model of TBI, providing encouraging results which support the potential of this approach to treat patients suffering from TBI. These findings justify ongoing research toward uncovering the mechanisms which underlie the functional improvements exhibited by hUCB + G-CSF combination therapy, thereby facilitating its safe and effect transition into the clinic. This paper is a review article. Referred literature in this paper has been listed in the reference section. The datasets supporting the conclusions of this article are available online by searching various databases, including PubMed. Some original points in this article come from the laboratory practice in our research center and the authors’ experiences.
Collapse
Affiliation(s)
- Michael G Liska
- Center of Excellence for Aging and Brain Repair, Tampa, FL 33612, USA
| | - Ike Dela Peña
- Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, College of Pharmacy, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
8
|
Wang WW, Han JH, Wang L, Bao TH. Scutellarin may alleviate cognitive deficits in a mouse model of hypoxia by promoting proliferation and neuronal differentiation of neural stem cells. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2017; 20:272-279. [PMID: 28392899 PMCID: PMC5378964 DOI: 10.22038/ijbms.2017.8355] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Objective(s): Scutellarin, a flavonoid extracted from the medicinal herb Erigeron breviscapus Hand-Mazz, protects neurons from damage and inhibits glial activation. Here we examined whether scutellarin may also protect neurons from hypoxia-induced damage. Materials and Methods: Mice were exposed to hypoxia for 7 days and then administered scutellarin (50 mg/kg/d) or vehicle for 30 days Cognitive impairment in the two groups was assessed using the Morris water maze test, cell proliferation in the hippocampus was compared using 5-bromo-2-deoxyuridine (BrdU) immunohistochemistry, and hippocampal levels of nestin and neuronal class III β-tubulin (Tuj-1) were measured using Western blotting. These results were validated in vitro by treating cultured neural stem cells (NSCs) with scutellarin (30 μM). Results: Treating mice with scutellarin shortened escape times and increased the number of platform crossings, it increased the number of BrdU-positive proliferating cells in the hippocampus, and it up-regulated expression of nestin and Tuj-1. Treating NSC cultures with scutellarin increased the number of proliferating cells and the proportion of cells differentiating into neurons instead of astrocytes. The increase in NSC proliferation was associated with phosphorylation of extracellular signal-regulated kinase (ERK) 1/2, while neuronal differentiation was associated with altered expression of differentiation-related genes. Conclusion: Scutellarin may alleviate cognitive impairment in a mouse model of hypoxia by promo-ting proliferation and neuronal differentiation of NSCs.
Collapse
Affiliation(s)
- Wei-Wei Wang
- Department of Cardiology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, PR China; Key Laboratory of Stem Cells and Regenerative Medicine, Institute of Molecular and Clinical Medicine, Kunming Medical University, Kunming, Yunnan, PR China; Department of Anatomy and Development Biology, Monash University, Clayton, vic 3800, Australia
| | - Jian-Hong Han
- The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, PR China
| | - Lin Wang
- The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, PR China
| | - Tian-Hao Bao
- The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, PR China; Mental Health Center of Kunming Medical University, Kunming City, Yunnan Province, PR China
| |
Collapse
|
9
|
De La Peña I, Sanberg PR, Acosta S, Lin SZ, Borlongan CV. G-CSF as an adjunctive therapy with umbilical cord blood cell transplantation for traumatic brain injury. Cell Transplant 2015; 24:447-57. [PMID: 25646620 DOI: 10.3727/096368915x686913] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Traumatic brain injury (TBI), a major contributor to deaths and permanent disability worldwide, has been recently described as a progressive cell death process rather than an acute event. TBI pathophysiology is complicated and can be distinguished by the initial primary injury and the subsequent secondary injury that ensues days after the trauma. Therapeutic opportunities for TBI remain very limited with patients subjected to surgery or rehabilitation therapy. The efficacy of stem cell-based interventions, as well as neuroprotective agents in other neurological disorders of which pathologies overlap with TBI, indicates their potential as alternative TBI treatments. Furthermore, their therapeutic limitations may be augmented when combination therapy is pursued instead of using a single agent. Indeed, we demonstrated remarkable combined efficacy of human umbilical cord blood (hUCB) cell therapy and granulocyte-colony-stimulating factor (G-CSF) treatment in TBI models, providing essential evidence for the translation of this approach to treat TBI. Further studies are warranted to determine the mechanisms underlying therapeutic benefits exerted by hUCB + G-CSF in order to enhance its safety and efficacy in the clinic.
Collapse
Affiliation(s)
- Ike De La Peña
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | | | | | | | | |
Collapse
|
10
|
Medina BNSP, Santos de Abreu I, Cavalcante LA, Silva WAB, da Fonseca RN, Allodi S, de Barros CM. 3-acetylpyridine-induced degeneration in the adult ascidian neural complex: Reactive and regenerative changes in glia and blood cells. Dev Neurobiol 2014; 75:877-93. [PMID: 25484282 DOI: 10.1002/dneu.22255] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 12/02/2014] [Indexed: 11/07/2022]
Abstract
Ascidians are interesting neurobiological models because of their evolutionary position as a sister-group of vertebrates and the high regenerative capacity of their central nervous system (CNS). We investigated the degeneration and regeneration of the cerebral ganglion complex of the ascidian Styela plicata following injection of the niacinamide antagonist 3-acetylpyridine (3AP), described as targeting the CNS of several vertebrates. For the analysis and establishment of a new model in ascidians, the ganglion complex was dissected and prepared for transmission electron microscopy (TEM), routine light microscopy (LM), immunohistochemistry and Western blotting, 1 or 10 days after injection of 3AP. The siphon stimulation test (SST) was used to quantify the functional response. One day after the injection of 3AP, CNS degeneration and recruitment of a non-neural cell type to the site of injury was observed by both TEM and LM. Furthermore, weaker immunohistochemical reactions for astrocytic glial fibrillary acidic protein (GFAP) and neuronal βIII-tubulin were observed. In contrast, the expression of caspase-3, a protein involved in the apoptotic pathway, and the glycoprotein CD34, a marker for hematopoietic stem cells, increased. Ten days after the injection of 3AP, the expression of markers tended toward the original condition. The SST revealed attenuation and subsequent recovery of the reflexes from 1 to 10 days after 3AP. Therefore, we have developed a new method to study ascidian neural degeneration and regeneration, and identified the decreased expression of GFAP and recruitment of blood stem cells to the damaged ganglion as reasons for the success of neuroregeneration in ascidians.
Collapse
Affiliation(s)
- Bianca N S P Medina
- Laboratório Integrado de Morfologia, Núcleo em Ecologia e Desenvolvimento Sócio Ambiental de Macaé, NUPEM, Universidade Federal do Rio de Janeiro, UFRJ, Macaé, RJ, Brazil.,Laboratório de Neurobiologia Comparativa e do Desenvolvimento, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, RJ, Brazil.,Pós-graduação em Produtos Bioativos e Biociências, Universidade Federal do Rio de Janeiro, UFRJ, Macaé, RJ, Brazil.,Pós-graduação em Ciências Morfológicas, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, RJ, Brazil
| | - Isadora Santos de Abreu
- Laboratório de Neurobiologia Comparativa e do Desenvolvimento, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, RJ, Brazil.,Pós-graduação em Ciências Morfológicas, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, RJ, Brazil.,Pós-graduação em Ciências Biológicas-Fisiologia, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, RJ, Brazil
| | - Leny A Cavalcante
- Laboratório de Neurobiologia Comparativa e do Desenvolvimento, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, RJ, Brazil.,Pós-graduação em Ciências Biológicas-Fisiologia, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, RJ, Brazil
| | - Wagner A B Silva
- Laboratório de Neurobiologia Comparativa e do Desenvolvimento, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, RJ, Brazil.,Pós-graduação em Ciências Biológicas-Fisiologia, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, RJ, Brazil
| | - Rodrigo N da Fonseca
- Laboratório Integrado de Bioquímica Hatisaburo Masuda, Núcleo em Ecologia e Desenvolvimento Sócio Ambiental de Macaé, NUPEM, Universidade Federal do Rio de Janeiro, UFRJ, Macaé, RJ, Brazil.,Pós-graduação em Produtos Bioativos e Biociências, Universidade Federal do Rio de Janeiro, UFRJ, Macaé, RJ, Brazil
| | - Silvana Allodi
- Laboratório de Neurobiologia Comparativa e do Desenvolvimento, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, RJ, Brazil.,Pós-graduação em Ciências Morfológicas, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, RJ, Brazil.,Pós-graduação em Ciências Biológicas-Fisiologia, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, RJ, Brazil
| | - Cintia M de Barros
- Laboratório Integrado de Morfologia, Núcleo em Ecologia e Desenvolvimento Sócio Ambiental de Macaé, NUPEM, Universidade Federal do Rio de Janeiro, UFRJ, Macaé, RJ, Brazil.,Pós-graduação em Produtos Bioativos e Biociências, Universidade Federal do Rio de Janeiro, UFRJ, Macaé, RJ, Brazil
| |
Collapse
|
11
|
Repair of spinal cord injury by inhibition of astrocyte growth and inflammatory factor synthesis through local delivery of flavopiridol in PLGA nanoparticles. Biomaterials 2014; 35:6585-94. [DOI: 10.1016/j.biomaterials.2014.04.042] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 04/14/2014] [Indexed: 12/30/2022]
|
12
|
Terashima T, Kojima H, Urabe H, Yamakawa I, Ogawa N, Kawai H, Chan L, Maegawa H. Stem cell factor-activated bone marrow ameliorates amyotrophic lateral sclerosis by promoting protective microglial migration. J Neurosci Res 2014; 92:856-69. [PMID: 24936617 PMCID: PMC4061499 DOI: 10.1002/jnr.23368] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive disease associated with motor neuron death. Several experimental treatments, including cell therapy using hematopoietic or neuronal stem cells, have been tested in ALS animal models, but therapeutic benefits have been modest. Here we used a new therapeutic strategy, bone marrow transplantation (BMT) with stem cell factor (SCF)- or FMS-like tyrosine kinase 3 (flt3)-activated bone marrow (BM) cells for the treatment of hSOD1(G93A) transgenic mice. Motor function and survival showed greater improvement in the SCF group than in the group receiving BM cells that had not been activated (BMT alone group), although no improvement was shown in the flt3 group. In addition, larger numbers of BM-derived cells that expressed the microglia marker Iba1 migrated to the spinal cords of recipient mice compared with the BMT alone group. Moreover, after SCF activation, but not flt3 activation or no activation, the migrating microglia expressed glutamate transporter-1 (GLT-1). In spinal cords in the SCF group, inflammatory cytokines tumor necrosis factor-α and interleukin-1β were suppressed and the neuroprotective molecule insulin-like growth factor-1 increased relative to nontreatment hSOD1(G93A) transgenic mice. Therefore, SCF activation changed the character of the migrating donor BM cells, which resulted in neuroprotective effects. These studies have identified SCF-activated BM cells as a potential new therapeutic agent for the treatment of ALS.
Collapse
Affiliation(s)
- Tomoya Terashima
- Department of Medicine, Shiga University of Medical Science, Seta, Otsu, Shiga, Japan
| | - Hideto Kojima
- Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Seta, Otsu, Shiga, Japan
| | - Hiroshi Urabe
- Department of Medicine, Shiga University of Medical Science, Seta, Otsu, Shiga, Japan
| | - Isamu Yamakawa
- Department of Medicine, Shiga University of Medical Science, Seta, Otsu, Shiga, Japan
| | - Nobuhiro Ogawa
- Department of Medicine, Shiga University of Medical Science, Seta, Otsu, Shiga, Japan
| | - Hiromichi Kawai
- Department of Medicine, Shiga University of Medical Science, Seta, Otsu, Shiga, Japan
| | - Lawrence Chan
- Departments of Medicine and Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Hiroshi Maegawa
- Department of Medicine, Shiga University of Medical Science, Seta, Otsu, Shiga, Japan
| |
Collapse
|
13
|
Kanski R, van Strien ME, van Tijn P, Hol EM. A star is born: new insights into the mechanism of astrogenesis. Cell Mol Life Sci 2014; 71:433-47. [PMID: 23907612 PMCID: PMC11113452 DOI: 10.1007/s00018-013-1435-9] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Revised: 07/10/2013] [Accepted: 07/22/2013] [Indexed: 12/22/2022]
Abstract
Astrocytes emerge as crucial cells for proper neuronal functioning in the developing and adult brain. Neurons and astrocytes are sequentially generated from the same pool of neural stem cells (NSCs). Tight regulation of the neuron-to-astrocyte switch is critical for (1) the generation of a balanced number of astrocytes and neurons and (2) neuronal circuit formation, since newborn astrocytes regulate synapse formation. This review focuses on signaling pathways that instruct astrogenesis, incorporating recently discovered intrinsic and extrinsic regulators. The canonical pathway of astrocytic gene expression, JAK/STAT signaling, is inhibited during neurogenesis to prevent premature astrocyte differentiation. At the onset of astrogenesis, Notch signaling induces epigenetic remodeling of astrocytic genes like glial fibrillary acidic protein to change NSC competence. In turn, astrogenesis is initiated by signals received from newborn neurons. We highlight how key molecular pathways like JAK/STAT and Notch are integrated in a complex network of environmental signals and epigenetic and transcriptional regulators to determine NSC differentiation. It is essential to understand NSC differentiation in respect to future NSC-based therapies for brain diseases, as transplanted NSCs preferentially become astrocytes. As emphasized in this review, many clues in this respect can be learned from development.
Collapse
Affiliation(s)
- Regina Kanski
- Astrocyte Biology and Neurodegeneration, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands
| | - Miriam E. van Strien
- Astrocyte Biology and Neurodegeneration, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands
| | - Paula van Tijn
- Astrocyte Biology and Neurodegeneration, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands
- Hubrecht Institute, an Institute of the Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands
| | - Elly M. Hol
- Astrocyte Biology and Neurodegeneration, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
14
|
Wang L, Duan D, Zhao Z, Teng X, Ge L, Liu B, Lu M. Repair of spinal cord injury by hypoxia-inducible factor-1a-expressing neural stem cells. JOURNAL OF MEDICAL HYPOTHESES AND IDEAS 2014. [DOI: 10.1016/j.jmhi.2013.07.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
15
|
Su Y, Cui L, Piao C, Li B, Zhao LR. The effects of hematopoietic growth factors on neurite outgrowth. PLoS One 2013; 8:e75562. [PMID: 24116056 PMCID: PMC3792965 DOI: 10.1371/journal.pone.0075562] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 08/14/2013] [Indexed: 01/06/2023] Open
Abstract
Stem cell factor (SCF) and granulocyte colony-stimulating factor (G-CSF) are initially discovered as the essential hematopoietic growth factors regulating bone marrow stem cell proliferation and differentiation, and SCF in combination with G-CSF (SCF+G-CSF) has synergistic effects on bone marrow stem cell mobilization. In this study we have determined the effect of SCF and G-CSF on neurite outgrowth in rat cortical neurons. Using molecular and cellular biology and live cell imaging approaches, we have revealed that receptors for SCF and G-CSF are expressed on the growth core of cortical neurons, and that SCF+G-CSF synergistically enhances neurite extension through PI3K/AKT and NFκB signaling pathways. Moreover, SCF+G-CSF induces much greater NFκB activation, NFκB transcriptional binding and brain-derived neurotrophic factor (BDNF) production than SCF or G-CSF alone. In addition, we have also observed that BDNF, the target gene of NFκB, is required for SCF+G-CSF-induced neurite outgrowth. These data suggest that SCF+G-CSF has synergistic effects to promote neurite growth. This study provides new insights into the contribution of hematopoietic growth factors in neuronal plasticity.
Collapse
Affiliation(s)
- Ye Su
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, Louisiana, United States of America
| | - Lili Cui
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, Louisiana, United States of America
- Department of Neurosurgery, State University of New York Upstate Medical University, Syracuse, New York, United States of America
| | - Chunshu Piao
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, Louisiana, United States of America
| | - Bin Li
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, Louisiana, United States of America
| | - Li-Ru Zhao
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, Louisiana, United States of America
- Department of Neurosurgery, State University of New York Upstate Medical University, Syracuse, New York, United States of America
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, Louisiana, United States of America
- * E-mail:
| |
Collapse
|
16
|
Sypecka J, Sarnowska A. The neuroprotective effect exerted by oligodendroglial progenitors on ischemically impaired hippocampal cells. Mol Neurobiol 2013; 49:685-701. [PMID: 24085562 PMCID: PMC3950613 DOI: 10.1007/s12035-013-8549-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 08/27/2013] [Indexed: 01/13/2023]
Abstract
Oligodendrocyte progenitor cells (OPCs) are the focus of intense research for the purpose of cell replacement therapies in acquired or inherited neurodegenerative disorders, accompanied by ongoing hypo/demyelination. Recently, it has been postulated that these glia-committed cells exhibit certain properties of neural stem cells. Advances in stem cell biology have shown that their therapeutic effect could be attributed to their ability to secret numerous active compounds which modify the local microenvironment making it more susceptible to restorative processes. To verify this hypothesis, we set up an ex vivo co-culture system of OPCs isolated from neonatal rat brain with organotypic hippocampal slices (OHC) injured by oxygen-glucose deprivation (OGD). The presence of OPCs in such co-cultures resulted in a significant neuroprotective effect manifesting itself as a decrease in cell death rate and as an extension of newly formed cells in ischemically impaired hippocampal slices. A microarray analysis of broad spectrum of trophic factors and cytokines expressed by OPCs was performed for the purpose of finding the factor(s) contributing to the observed effect. Three of them—BDNF, IL-10 and SCF—were selected for the subsequent functional assays. Our data revealed that BDNF released by OPCs is the potent factor that stimulates cell proliferation and survival in OHC subjected to OGD injury. At the same time, it was observed that IL-10 attenuates inflammatory processes by promoting the formation of the cells associated with the immunological response. Those neuroprotective qualities of oligodendroglia-biased progenitors significantly contribute to anticipating a successful cell replacement therapy.
Collapse
Affiliation(s)
- Joanna Sypecka
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5, Pawinskiego str.,, 02-106, Warsaw, Poland,
| | | |
Collapse
|
17
|
Velkey JM, O'Shea KS. Expression of Neurogenin 1 in mouse embryonic stem cells directs the differentiation of neuronal precursors and identifies unique patterns of down-stream gene expression. Dev Dyn 2013; 242:230-53. [PMID: 23288605 DOI: 10.1002/dvdy.23920] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Revised: 11/16/2012] [Accepted: 11/16/2012] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Delineating the cascades of growth and transcription factor expression that shape the developing nervous system will improve our understanding of its molecular histogenesis and suggest strategies for cell replacement therapies. In the current investigation, we examined the ability of the proneural gene, Neurogenin1 (Neurog1; also Ngn1, Neurod3), to drive differentiation of pluripotent embryonic stem cells (ESC). RESULTS Transient expression of Neurog1 in ESC was sufficient to initiate neuronal differentiation, and produced neuronal subtypes reflecting its expression pattern in vivo. To begin to address the molecular mechanisms involved, we used microarray analysis to identify potential down-stream targets of Neurog1 expressed at sequential stages of neuronal differentiation. CONCLUSIONS ESC expressing Neurogenin1 begin to withdraw from cycle and form precursors that differentiate exclusively into neurons. This work identifies unique patterns of gene expression following expression of Neurog1, including genes and signaling pathways involved in process outgrowth and cell migration, regional differentiation of the nervous system, and cell cycle.
Collapse
Affiliation(s)
- J Matthew Velkey
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | | |
Collapse
|