1
|
Wu J, Li R, Wang J, Zhu H, Ma Y, You C, Shu K. Reactive Astrocytes in Glioma: Emerging Opportunities and Challenges. Int J Mol Sci 2025; 26:2907. [PMID: 40243478 PMCID: PMC11989224 DOI: 10.3390/ijms26072907] [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: 02/14/2025] [Revised: 03/16/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Gliomas are the most prevalent malignant tumors in the adult central nervous system (CNS). Glioblastoma (GBM) accounts for approximately 60-70% of primary gliomas. It is a great challenge to human health because of its high degree of malignancy, rapid progression, short survival time, and susceptibility to recurrence. Owing to the specificity of the CNS, the glioma microenvironment often contains numerous glial cells. Astrocytes are most widely distributed in the human brain and form reactive astrocyte proliferation regions around glioma tissue. In addition, astrocytes are activated under pathological conditions and regulate tumor and microenvironmental cells through cell-to-cell contact or the secretion of active substances. Therefore, astrocytes have attracted attention as important components of the glioma microenvironment. Here, we focus on the mechanisms of reactive astrocyte activation under glioma conditions, their contribution to the mechanisms of glioma genesis and progression, and their potential value as targets for clinical intervention in gliomas.
Collapse
Affiliation(s)
| | | | | | | | | | - Chao You
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095, Jie Fang Avenue, Qiao Kou District, Wuhan 430030, China; (J.W.); (J.W.); (H.Z.); (Y.M.)
| | - Kai Shu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095, Jie Fang Avenue, Qiao Kou District, Wuhan 430030, China; (J.W.); (J.W.); (H.Z.); (Y.M.)
| |
Collapse
|
2
|
Chang GQ, Yasmin N, Collier AD, Karatayev O, Khalizova N, Onoichenco A, Fam M, Albeg AS, Campbell S, Leibowitz SF. Fibroblast growth factor 2: Role in prenatal alcohol-induced stimulation of hypothalamic peptide neurons. Prog Neuropsychopharmacol Biol Psychiatry 2022; 116:110536. [PMID: 35176416 PMCID: PMC8920779 DOI: 10.1016/j.pnpbp.2022.110536] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 02/08/2022] [Accepted: 02/10/2022] [Indexed: 12/13/2022]
Abstract
Prenatal alcohol exposure (PAE) increases alcohol consumption and risk for alcohol use disorder. This phenomenon in rodents is suggested to involve a stimulatory effect of PAE, in female more than male offspring, on neurogenesis and density of neurons expressing neuropeptides in lateral hypothalamus (LH), including melanin-concentrating hormone (MCH), known to promote alcohol intake. With evidence suggesting a role for fibroblast growth factor 2 (FGF2) and its receptor FGFR1 in stimulating neurogenesis and alcohol drinking, we investigated here whether the FGF2-FGFR1 system is involved in the PAE-induced increase in MCH neurons, in postnatal offspring of pregnant rats given ethanol orally (embryonic day 10-15) at a low-moderate (2 g/kg/day) or high (5 g/kg/day) dose. Our results demonstrate that PAE at the low-moderate but not high dose stimulates FGF2 and FGFR1 gene expression and increases the density of MCH neurons co-expressing FGF2, only in females, but FGFR1 in both sexes. PAE induces this effect in the dorsal but not ventral area of the LH. Further analysis of FGF2 and FGFR1 transcripts within individual MCH neurons reveals an intracellular, sex-dependent effect, with PAE increasing FGF2 transcripts positively related to FGFR1 in the nucleus as well as cytoplasm of females but transcripts only in the cytoplasm of males. Peripheral injection of FGF2 itself (80 μg/kg, s.c.) in pregnant rats mimics these effects of PAE. Together, these results support the involvement of the FGF2-FGFR1 system in mediating the PAE-induced, sex dependent increase in density of MCH neurons, possibly contributing to increased alcohol consumption in the offspring.
Collapse
Affiliation(s)
- Guo-Qing Chang
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065, United States of America
| | - Nushrat Yasmin
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065, United States of America
| | - Adam D Collier
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065, United States of America
| | - Olga Karatayev
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065, United States of America
| | - Nailya Khalizova
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065, United States of America
| | - Amanda Onoichenco
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065, United States of America
| | - Milisia Fam
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065, United States of America
| | - Avi S Albeg
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065, United States of America
| | - Samantha Campbell
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065, United States of America
| | - Sarah F Leibowitz
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065, United States of America.
| |
Collapse
|
3
|
Puebla M, Tapia PJ, Espinoza H. Key Role of Astrocytes in Postnatal Brain and Retinal Angiogenesis. Int J Mol Sci 2022; 23:ijms23052646. [PMID: 35269788 PMCID: PMC8910249 DOI: 10.3390/ijms23052646] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 02/21/2022] [Accepted: 02/23/2022] [Indexed: 01/27/2023] Open
Abstract
Angiogenesis is a key process in various physiological and pathological conditions in the nervous system and in the retina during postnatal life. Although an increasing number of studies have addressed the role of endothelial cells in this event, the astrocytes contribution in angiogenesis has received less attention. This review is focused on the role of astrocytes as a scaffold and in the stabilization of the new blood vessels, through different molecules release, which can modulate the angiogenesis process in the brain and in the retina. Further, differences in the astrocytes phenotype are addressed in glioblastoma, one of the most devastating types of brain cancer, in order to provide potential targets involved in the cross signaling between endothelial cells, astrocytes and glioma cells, that mediate tumor progression and pathological angiogenesis. Given the relevance of astrocytes in angiogenesis in physiological and pathological conditions, future studies are required to better understand the interrelation between endothelial and astrocyte signaling pathways during this process.
Collapse
Affiliation(s)
- Mariela Puebla
- Centro de Fisiología Celular e Integrativa, Facultad de Medicina-Clínica Alemana, Universidad del Desarrollo, Av. Plaza 680, Las Condes, Santiago 7550000, Chile;
| | - Pablo J. Tapia
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Av. Lota 2465, Providencia, Santiago 7500000, Chile;
- Facultad de Medicina Veterinaria y Agronomía, Universidad de las Américas, Av. República 71, Santiago 8320000, Chile
| | - Hilda Espinoza
- Facultad de Ciencias de la Salud, Universidad del Alba, Av. Ejército Libertador 171, Santiago 8320000, Chile
- Correspondence:
| |
Collapse
|
4
|
Xia J, Xue X, Liu W, Qi Z, Liu W. The Role of Fgf9 in the Antidepressant Effects of Exercise and Fluoxetine in Chronic Unpredictable Mild Stress Mice. Psychosom Med 2021; 83:795-804. [PMID: 33938506 DOI: 10.1097/psy.0000000000000953] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
OBJECTIVE The neurotrophic hypothesis of depression posits that stress and depression decrease neurotrophic factor expression in brain, whereas antidepressants and exercise can contribute to the blockade of stress effects and produce antidepressant effects. Fibroblast growth factor 9 (FGF9), a member of the fibroblast growth factor (FGF) family, has been reported to be dysregulated in depression. The present study aimed to determine whether and how Fgf9 mediates the antidepressant effects of fluoxetine and exercise in chronic unpredictable mild stress (CUMS) mice. METHODS Male C57BL/6 mice were exposed to CUMS for 7 weeks. From the fourth week, CUMS-exposed mice were subjected to fluoxetine treatment or swimming exercise for 4 weeks. Forced swim test, tail suspension test, and hole-board test were used to assess behaviors of mice. Real-time polymerase chain reaction was used to examine hippocampal messenger RNA levels of Fgf9, Fgf2, FgfR1, FgfR2, and FgfR3. Western blotting was used to examine the protein levels of Fgf9, protein kinase B (Akt), and phosphorylation of Akt at Ser473 in mouse hippocampus. RESULTS Our results demonstrated that CUMS induced depression-like behaviors, which were reversed by fluoxetine treatment and swimming exercise. Moreover, we found that CUMS resulted in a dysregulation of Fgf9, Fgf2, and FgfR2 expression, whereas fluoxetine and swimming restored the FGF expression in CUMS-exposed mice. An analysis of the proteins suggests that the antidepressant effects of fluoxetine and exercise in CUMS-exposed mice were associated with ameliorated Fgf9/Akt signaling. CONCLUSIONS Our findings have demonstrated that swimming exercise mimics the antidepressant effects of fluoxetine by regulating Fgf9 in CUMS-exposed mice, which may offer new mechanism-based therapeutic targets for depression.
Collapse
Affiliation(s)
- Jie Xia
- From the Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education (Xia, Xue, Wenbin Liu, Qi, Weina Liu), College of Physical Education and Health (Xia, Wenbin Liu, Qi, Weina Liu), East China Normal University; and Key Laboratory of Exercise and Health Sciences of Ministry of Education (Xue), Shanghai University of Sport, Shanghai, China
| | | | | | | | | |
Collapse
|
5
|
Caffino L, Mottarlini F, Zita G, Gawliński D, Gawlińska K, Wydra K, Przegaliński E, Fumagalli F. The effects of cocaine exposure in adolescence: Behavioural effects and neuroplastic mechanisms in experimental models. Br J Pharmacol 2021; 179:4233-4253. [PMID: 33963539 PMCID: PMC9545182 DOI: 10.1111/bph.15523] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 04/15/2021] [Accepted: 05/04/2021] [Indexed: 01/23/2023] Open
Abstract
Drug addiction is a devastating disorder with a huge economic and social burden for modern society. Although an individual may slip into drug abuse throughout his/her life, adolescents are at higher risk, but, so far, only a few studies have attempted to elucidate the underlying cellular and molecular bases of such vulnerability. Indeed, preclinical evidence indicates that psychostimulants and adolescence interact and contribute to promoting a dysfunctional brain. In this review, we have focused our attention primarily on changes in neuroplasticity brought about by cocaine, taking into account that there is much less evidence from exposure to cocaine in adolescence, compared with that from adults. This review clearly shows that exposure to cocaine during adolescence, acute or chronic, as well as contingent or non‐contingent, confers a vulnerable endophenotype, primarily, by causing changes in neuroplasticity. Given the close relationship between drug abuse and psychiatric disorders, we also discuss the translational implications providing an interpretative framework for clinical studies involving addictive as well as affective or psychotic behaviours.
Collapse
Affiliation(s)
- Lucia Caffino
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Francesca Mottarlini
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Gianmaria Zita
- Dipartimento di Salute Mentale e Dipendenze, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Dawid Gawliński
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Kraków, Poland
| | - Kinga Gawlińska
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Kraków, Poland
| | - Karolina Wydra
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Kraków, Poland
| | - Edmund Przegaliński
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Kraków, Poland
| | - Fabio Fumagalli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
6
|
Yao Z, Zhang Z, Zhang J, Cai X, Zhong Z, Huang Y, Qu S. Electroacupuncture alleviated the depression-like behavior by regulating FGF2 and astrocytes in the hippocampus of rats with chronic unpredictable mild stress. Brain Res Bull 2021; 169:43-50. [PMID: 33434624 DOI: 10.1016/j.brainresbull.2021.01.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 11/24/2020] [Accepted: 01/05/2021] [Indexed: 12/16/2022]
Abstract
Studies have shown that basic fibroblast growth factor (FGF2) is a neurotrophic factor associated with depression. Electroacupuncture (EA) has been shown to be an effective treatment for depression. In the current study, we observed the effects of EA on hippocampal FGF2 and astrocytes, and further investigated the mechanism underlying antidepressant effect of EA. The chronic unpredictable mild stress (CUMS) method were selected to induce depressive-like behaviors of rats. Paroxetine is a commonly used antidepressant and was used as a positive control drug in this experiment. The male adult Sprague Dawley (SD) rats were randomized to four experimental groups (normal control group, CUMS group, EA group and paroxetine group, n = 10/group). EA intervention was administered once daily for 14 days at acupuncture points Baihui (GV20) and Yintang (GV29). Rats in the paroxetine group received daily paroxetine administered intragastrical. Behavioral test, immunohistochemistry (IHC), western blot (WB) and quantitative real-time PCR (qPCR) were conducted to evaluate the intervene effect and the changes of FGF2 and astrocyte marker (glial fibrillary acidic protein, GFAP). The results showed that EA and paroxetine could improve depression-like behavior in CUMS rats, and up-regulated the expression level of FGF2 in the hippocampus, increased GFAP protein expression and the mean optical density of GFAP-immunoreactive astrocyte (GFAP-ir astrocyte). Our findings have identified that EA could ameliorate depressive-like behaviors possibly by regulating the expression of FGF2 in the hippocampus, and the mechanism might be related to the effect of FGF2 on astrocytes.
Collapse
Affiliation(s)
- Zengyu Yao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
| | - Zhinan Zhang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
| | - Jiping Zhang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
| | - Xiaowen Cai
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
| | - Zheng Zhong
- Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
| | - Yong Huang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong Province, 510515, China.
| | - Shanshan Qu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong Province, 510515, China.
| |
Collapse
|
7
|
Neve A, Migliavacca J, Capdeville C, Schönholzer MT, Gries A, Ma M, Santhana Kumar K, Grotzer M, Baumgartner M. Crosstalk between SHH and FGFR Signaling Pathways Controls Tissue Invasion in Medulloblastoma. Cancers (Basel) 2019; 11:cancers11121985. [PMID: 31835472 PMCID: PMC6966681 DOI: 10.3390/cancers11121985] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/25/2019] [Accepted: 12/06/2019] [Indexed: 12/13/2022] Open
Abstract
In the Sonic Hedgehog (SHH) subgroup of medulloblastoma (MB), tumor initiation and progression are in part driven by smoothened (SMO) and fibroblast growth factor (FGF)-receptor (FGFR) signaling, respectively. We investigated the impact of the SMO-FGFR crosstalk on tumor growth and invasiveness in MB. We found that FGFR signaling represses GLI1 expression downstream of activated SMO in the SHH MB line DAOY and induces MKI67, HES1, and BMI1 in DAOY and in the group 3 MB line HD-MBO3. FGFR repression of GLI1 does not affect proliferation or viability, whereas inhibition of FGFR is necessary to release SMO-driven invasiveness. Conversely, SMO activation represses FGFR-driven sustained activation of nuclear ERK. Parallel activation of FGFR and SMO in ex vivo tumor cell-cerebellum slice co-cultures reduced invasion of tumor cells without affecting proliferation. In contrast, treatment of the cells with the SMO antagonist Sonidegib (LDE225) blocked invasion and proliferation in cerebellar slices. Thus, sustained, low-level SMO activation is necessary for proliferation and tissue invasion, whereas acute, pronounced activation of SMO can repress FGFR-driven invasiveness. This suggests that the tumor cell response is dependent on the relative local abundance of the two factors and indicates a paradigm of microenvironmental control of invasion in SHH MB through mutual control of SHH and FGFR signaling.
Collapse
Affiliation(s)
- Anuja Neve
- Department of Oncology, University Children’s Hospital Zürich, CH-8032 Zürich, Switzerland; (A.N.); (J.M.); (C.C.); (M.T.S.); (A.G.); (K.S.K.); (M.G.)
| | - Jessica Migliavacca
- Department of Oncology, University Children’s Hospital Zürich, CH-8032 Zürich, Switzerland; (A.N.); (J.M.); (C.C.); (M.T.S.); (A.G.); (K.S.K.); (M.G.)
| | - Charles Capdeville
- Department of Oncology, University Children’s Hospital Zürich, CH-8032 Zürich, Switzerland; (A.N.); (J.M.); (C.C.); (M.T.S.); (A.G.); (K.S.K.); (M.G.)
| | - Marc Thomas Schönholzer
- Department of Oncology, University Children’s Hospital Zürich, CH-8032 Zürich, Switzerland; (A.N.); (J.M.); (C.C.); (M.T.S.); (A.G.); (K.S.K.); (M.G.)
| | - Alexandre Gries
- Department of Oncology, University Children’s Hospital Zürich, CH-8032 Zürich, Switzerland; (A.N.); (J.M.); (C.C.); (M.T.S.); (A.G.); (K.S.K.); (M.G.)
| | - Min Ma
- Faculty of Biology and Medicine, University of Lausanne, Biochemistry, CH-1066 Epalinges, Switzerland;
| | - Karthiga Santhana Kumar
- Department of Oncology, University Children’s Hospital Zürich, CH-8032 Zürich, Switzerland; (A.N.); (J.M.); (C.C.); (M.T.S.); (A.G.); (K.S.K.); (M.G.)
| | - Michael Grotzer
- Department of Oncology, University Children’s Hospital Zürich, CH-8032 Zürich, Switzerland; (A.N.); (J.M.); (C.C.); (M.T.S.); (A.G.); (K.S.K.); (M.G.)
| | - Martin Baumgartner
- Department of Oncology, University Children’s Hospital Zürich, CH-8032 Zürich, Switzerland; (A.N.); (J.M.); (C.C.); (M.T.S.); (A.G.); (K.S.K.); (M.G.)
- Correspondence: ; Tel.: +41-44-266-3730
| |
Collapse
|
8
|
Wu HE, Teixeira AL, Barroso L, Silva APM, de Souza Nicolau M, Ferreira JDR, Bertola L, Vieira EM, Diniz BS. Epidermal growth factor and fibroblast growth factor-2 circulating levels in elderly with major depressive disorder. Psychiatry Res 2019; 272:141-143. [PMID: 30583255 DOI: 10.1016/j.psychres.2018.12.084] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 12/13/2018] [Accepted: 12/16/2018] [Indexed: 12/31/2022]
Abstract
Epidermal growth factor (EGF) and Fibroblast Growth Factor-2 (FGF-2) are growth factors involved neuronal growth and synaptic plasticity. These markers have been implicated in neuropsychiatric disorders, including major depression. However, no particular studies of EGF and FGF-2 have been conducted in older adults with major depressive disorder (MDD). In this study, we aim to investigate the plasma levels of EGF and FGF-2 in elderly with MDD. We included 89 older adults with MDD and 51 older (healthy control, HC) adults. The cognitive performance was evaluated by the Mattis Dementia Rating Scale (MDRS). The EGF and FGF-2 were measured by using multiplex assay for LUMINEX platform. There were also no significant differences between the patient group in terms of plasma levels of EGF and FGF-2 when compared to the HC group. There were not any significant correlations between plasma levels of EGF or FGF2 and MDRS total or individual scores in patient group and HC. There were significant correlations between plasma levels of EGF and FGF2 in both patient group and HC. Further study on plasma levels of EGF and FGF2 should be implemented in larger samples in elderly with MDD.
Collapse
Affiliation(s)
- Hanjing Emily Wu
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Antonio L Teixeira
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Lucelia Barroso
- Laboratory of Interdisciplinary Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais, Brazil
| | - Ana Paula Mendes Silva
- Laboratory of Interdisciplinary Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais, Brazil
| | - Mariana de Souza Nicolau
- Laboratory of Interdisciplinary Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais, Brazil
| | | | - Laiss Bertola
- Laboratory of Interdisciplinary Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais, Brazil
| | - Erica Marciano Vieira
- Laboratory of Interdisciplinary Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais, Brazil
| | - Breno Satler Diniz
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA; Department of Psychiatry, Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Center for Addiction and Mental Health (CAMH), Toronto, ON, Canada.
| |
Collapse
|
9
|
Zhou Y, Wang Z, Li J, Li X, Xiao J. Fibroblast growth factors in the management of spinal cord injury. J Cell Mol Med 2017; 22:25-37. [PMID: 29063730 PMCID: PMC5742738 DOI: 10.1111/jcmm.13353] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 07/12/2017] [Indexed: 12/11/2022] Open
Abstract
Spinal cord injury (SCI) possesses a significant health and economic burden worldwide. Traumatic SCI is a devastating condition that evolves through two successive stages. Throughout each of these stages, disturbances in ionic homeostasis, local oedema, ischaemia, focal haemorrhage, free radicals stress and inflammatory response were observed. Although there are no fully restorative cures available for SCI patients, various molecular, cellular and rehabilitative therapies, such as limiting local inflammation, preventing secondary cell death and enhancing the plasticity of local circuits in the spinal cord, were described. Current preclinical studies have showed that fibroblast growth factors (FGFs) alone or combination therapies utilizing cell transplantation and biomaterial scaffolds are proven effective for treating SCI in animal models. More importantly, some studies further demonstrated a paucity of clinical transfer usage to promote functional recovery of numerous patients with SCI. In this review, we focus on the therapeutic capacity and pitfalls of the FGF family and its clinical application for treating SCI, including the signalling component of the FGF pathway and the role in the central nervous system, the pathophysiology of SCI and the targets for FGF treatment. We also discuss the challenges and potential for the clinical translation of FGF-based approaches into treatments for SCI.
Collapse
Affiliation(s)
- Yulong Zhou
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.,Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhouguang Wang
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jiawei Li
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.,Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaokun Li
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jian Xiao
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.,Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
10
|
Wu CK, Tseng PT, Chen YW, Tu KY, Lin PY. Significantly higher peripheral fibroblast growth factor-2 levels in patients with major depressive disorder: A preliminary meta-analysis under MOOSE guidelines. Medicine (Baltimore) 2016; 95:e4563. [PMID: 27537581 PMCID: PMC5370807 DOI: 10.1097/md.0000000000004563] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND In vivo and in vitro studies demonstrate the important roles of fibroblast growth factor (FGF) and FGF receptors (FGFRs) in neural survival, neurogenesis, oxidative stress, and emotional behavior. However, evidence on the role of FGF and FGFR in the pathophysiology of major depressive disorder (MDD) remains limited and inconclusive. OBJECTIVES This preliminary meta-analysis aimed to examine changes in peripheral or central FGF and FGFR levels in patients with MDD. DATA SOURCES Electronic research through platform of PubMed and ClinicalTrials.gov. STUDY ELIGIBILITY CRITERIA, PARTICIPANTS, AND INTERVENTIONS We used the inclusion criteria: articles discussing the comparisons of FGF levels, either in peripheral or central environment, in patients with MDD and in healthy controls (HC); articles on clinical trials in humans; and case-control trials. Case reports or series and nonclinical trials were excluded. STUDY APPRAISAL AND SYNTHESIS METHODS Using a thorough literature search, the FGF/FGFR levels in patients with MDD and HC were compared. Four studies on peripheral FGF-2 and 3 on central FGF-2 and FGFR1 levels were included. RESULTS The findings reveal significantly higher peripheral FGF-2 protein and central FGFR1 RNA levels in patients with MDD than in HC (P = 0.005 and 0.006, separately), but no significant association with clinical variables. There was also no significant difference in the central FGF-2 levels in patients with MDD and in HC (P = 0.180). LIMITATION The study has limitations of a small number of included studies, lack of meta-analysis of the FGF changes along with treatment, and lack of direct evidence on correlation of peripheral FGF-2 with central FGF-2 levels. CONCLUSIONS AND IMPLICATIONS OF KEY FINDINGS This preliminary meta-analysis points out a new direction for future studies investigating the relationship among MDD, oxidative stress, and the FGF family.
Collapse
Affiliation(s)
- Ching-Kuan Wu
- Department of Psychiatry, Tsyr-Huey Mental Hospital, Kaohsiung Jen-Ai's Home
| | - Ping-Tao Tseng
- Department of Psychiatry, Tsyr-Huey Mental Hospital, Kaohsiung Jen-Ai's Home
| | | | - Kun-Yu Tu
- Department of Psychiatry, Tsyr-Huey Mental Hospital, Kaohsiung Jen-Ai's Home
| | - Pao-Yen Lin
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine
- Institute for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Correspondence: Pao-Yen Lin, Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital, 123, Dapi Road, Niaosong, Kaohsiung 833, Taiwan (e-mail: )
| |
Collapse
|
11
|
Giannotti G, Caffino L, Malpighi C, Melfi S, Racagni G, Fumagalli F. A single exposure to cocaine during development elicits regionally-selective changes in basal basic Fibroblast Growth Factor (FGF-2) gene expression and alters the trophic response to a second injection. Psychopharmacology (Berl) 2015; 232:713-9. [PMID: 25124315 DOI: 10.1007/s00213-014-3708-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 07/29/2014] [Indexed: 12/19/2022]
Abstract
RATIONALE During adolescence, the brain is maturing and more sensitive to drugs of abuse that can influence its developmental trajectory. Recently, attention has been focused on basic fibroblast growth factor (FGF-2) given that its administration early in life enhances the acquisition of cocaine self-administration and sensitization at adulthood (Turner et al. (Pharmacol Biochem Behav 92:100-4, 2009), Clinton et al. (Pharmacol Biochem Behav103:6-17, 2012)). Additionally, we found that abstinence from adolescent cocaine exposure long lastingly dysregulates FGF-2 transcription (Giannotti et al. (Psychopharmacology (Berl) 225:553-60, 2013 ). OBJECTIVES The objectives of the study are to evaluate if (1) a single injection of cocaine (20 mg/kg) at postnatal day 35 alters FGF-2 messenger RNA (mRNA) levels and (2) the first injection influences the trophic response to a second injection (10 mg/kg) provided 24 h or 7 days later. RESULTS We found regional differences in the FGF-2 expression pattern as either the first or the second injection of cocaine by themselves upregulated FGF-2 mRNA in the medial prefrontal cortex and nucleus accumbens while downregulating it in the hippocampus. The first injection influences the trophic response of the second. Of note, 24 h after the first injection, accumbal and hippocampal FGF-2 changes produced by cocaine in saline-pretreated rats were prevented in cocaine-pretreated rats. Conversely, in the medial prefrontal cortex and hippocampus 7 days after the first injection, the cocaine-induced FGF-2 changes were modified by the subsequent exposure to the psychostimulant. CONCLUSIONS These findings show that a single cocaine injection is sufficient to produce enduring changes in the adolescent brain and indicate that early cocaine priming alters the mechanisms regulating the trophic response in a brain region-specific fashion.
Collapse
Affiliation(s)
- Giuseppe Giannotti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Via Balzaretti 9, 20133, Milano, Italy
| | | | | | | | | | | |
Collapse
|
12
|
Kang K, Lee SW, Han JE, Choi JW, Song MR. The complex morphology of reactive astrocytes controlled by fibroblast growth factor signaling. Glia 2014; 62:1328-44. [PMID: 24796693 DOI: 10.1002/glia.22684] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Revised: 03/19/2014] [Accepted: 04/17/2014] [Indexed: 12/12/2022]
Abstract
Astrocytes are the most abundant cell-type of the human brain and play a variety of roles in brain homeostasis and synaptic maturation, under normal conditions. However, astrocytes undergo dramatic pathological changes in response to brain injury, such as reactive gliosis and glial scar formation. Although abnormal hypertrophy and massive proliferation of astrocytes are obvious, the molecular identity and cues that dictate the structural changes in reactive astrocytes remain unclear. This study proposes that fibroblast growth factor (FGF) signaling is responsible for making astrocyte morphology more complex and hypertrophic in response to an inflammatory stimulus such as lipopolysaccharide. Primary astrocytes isolated from perinatal brains developed more branches in the presence of FGF8 or lesser branches in the presence of FGF2. Introduction of the constitutively active form of the FGF receptor 3 (caFGFR3) into the brain increases the structural complexity, with greater glial fibrillary acidic protein level in astrocytes, while overexpression of a dominant-negative form of FGFR3 (dnFGFR3) reduces it. Treatment of FGF8 facilitated the wound-healing process of primary astrocytes in vitro by changing their morphology, indicating that the FGF signal may control the responsiveness of astrocytes in injury conditions. Finally, the blockade of FGF signaling by introducing dnFGFR3 at the site of reactive gliosis reduces astrocyte branch formation and minimizes hypertrophic responses during reactive gliosis. Taken together, these results indicate that FGF8-FGFR3 signaling controls structural changes in astrocytes during reactive gliosis, under pathogenic conditions.
Collapse
Affiliation(s)
- Kyungjoon Kang
- School of Life Sciences, Bioimaging Research Center and Cell Dynamics Research Center, Gwangju Institute of Science and Technology, Oryong-dong, Buk-gu, Gwangju, 500-712, Republic of Korea
| | | | | | | | | |
Collapse
|
13
|
Annenkov A. Receptor tyrosine kinase (RTK) signalling in the control of neural stem and progenitor cell (NSPC) development. Mol Neurobiol 2013; 49:440-71. [PMID: 23982746 DOI: 10.1007/s12035-013-8532-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 08/09/2013] [Indexed: 01/04/2023]
Abstract
Important developmental responses are elicited in neural stem and progenitor cells (NSPC) by activation of the receptor tyrosine kinases (RTK), including the fibroblast growth factor receptors, epidermal growth factor receptor, platelet-derived growth factor receptors and insulin-like growth factor receptor (IGF1R). Signalling through these RTK is necessary and sufficient for driving a number of developmental processes in the central nervous system. Within each of the four RTK families discussed here, receptors are activated by sets of ligands that do not cross-activate receptors of the other three families, and therefore, their activation can be independently regulated by ligand availability. These RTK pathways converge on a conserved core of signalling molecules, but differences between the receptors in utilisation of signalling molecules and molecular adaptors for intracellular signal propagation become increasingly apparent. Intracellular inhibitors of RTK signalling are widely involved in the regulation of developmental signalling in NSPC and often determine developmental outcomes of RTK activation. In addition, cellular responses of NSPC to the activation of a given RTK may be significantly modulated by signal strength. Cellular propensity to respond also plays a role in developmental outcomes of RTK signalling. In combination, these mechanisms regulate the balance between NSPC maintenance and differentiation during development and in adulthood. Attribution of particular developmental responses of NSPC to specific pathways of RTK signalling becomes increasingly elucidated. Co-activation of several RTK in developing NSPC is common, and analysis of co-operation between their signalling pathways may advance knowledge of RTK role in NSPC development.
Collapse
Affiliation(s)
- Alexander Annenkov
- Bone and Joint Research Unit, William Harvey Research Institute, Bart's and The London School of Medicine, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK,
| |
Collapse
|
14
|
Astrocyte infiltration into injectable collagen-based hydrogels containing FGF-2 to treat spinal cord injury. Biomaterials 2013; 34:3591-602. [DOI: 10.1016/j.biomaterials.2012.12.050] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2012] [Accepted: 12/29/2012] [Indexed: 11/23/2022]
|
15
|
Giannotti G, Caffino L, Calabrese F, Racagni G, Fumagalli F. Dynamic modulation of basic Fibroblast Growth Factor (FGF-2) expression in the rat brain following repeated exposure to cocaine during adolescence. Psychopharmacology (Berl) 2013; 225:553-60. [PMID: 22895673 DOI: 10.1007/s00213-012-2840-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Accepted: 08/01/2012] [Indexed: 12/13/2022]
Abstract
RATIONALE Our study stems from four related lines of evidence: (1) FGF-2 is expressed in the developing brain; (2) psychostimulants modulate FGF-2 expression; (3) stress alters FGF-2 expression; and (4) exogenous administration of FGF-2 long-lastingly alters cocaine acquisition of self-administration. OBJECTIVES This research aims to study the effects of adolescent cocaine exposure on FGF-2 mRNA levels and its influence on the response to stress. MATERIALS AND METHODS Rats were treated subcutaneously with saline or cocaine from postnatal day (PND) 28 to PND 42, a period that roughly approximates adolescence in humans. At PND 45 and PND 90, rats were exposed to an acute stress. Real-time PCRs were performed on total RNA extracted from the prefrontal cortex, hippocampus, nucleus accumbens and striatum. RESULTS In the prefrontal cortex, repeated cocaine treatment during adolescence increased FGF-2 mRNA levels in PND 90 rats and altered its response to an acute stress in both PND 45 and PND 90 rats. In the hippocampus of PND 45 rats, we found an increase of FGF-2 mRNA levels following repeated cocaine administration. No changes in the trophic factor gene expression were found in the striatum and nucleus accumbens. CONCLUSIONS Our data show that cocaine exposure during adolescence alters FGF-2 mRNA levels throughout life in rat prefrontal cortex and modulates its response to an adverse event. These results point to FGF-2 as a potential molecular target through which exposure to cocaine early in life may dynamically and persistently alter brain homeostasis.
Collapse
Affiliation(s)
- Giuseppe Giannotti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Via Balzaretti 9, Milan, Italy
| | | | | | | | | |
Collapse
|
16
|
Turner CA, Watson SJ, Akil H. The fibroblast growth factor family: neuromodulation of affective behavior. Neuron 2012; 76:160-74. [PMID: 23040813 DOI: 10.1016/j.neuron.2012.08.037] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2012] [Indexed: 12/20/2022]
Abstract
In this review, we propose a broader view of the role of the fibroblast growth factor (FGF) family in modulating brain function. We suggest that some of the FGF ligands together with the FGF receptors are altered in individuals with affective disorder and modulate emotionality in animal models. Thus, we propose that members of the FGF family may be genetic predisposing factors for anxiety, depression, or substance abuse; that they play a key organizing role during early development but continue to play a central role in neuroplasticity in adulthood; and that they work not only over extended time frames, but also via rapid signaling mechanisms, allowing them to exert an "on-line" influence on behavior. Therefore, the FGF family appears to be a prototype of "switch genes" that are endowed with organizational and modulatory properties across the lifespan, and that may represent molecular candidates as biomarkers and treatment targets for affective and addictive disorders.
Collapse
Affiliation(s)
- Cortney A Turner
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, USA.
| | | | | |
Collapse
|
17
|
Kolb B, Mychasiuk R, Muhammad A, Li Y, Frost DO, Gibb R. Experience and the developing prefrontal cortex. Proc Natl Acad Sci U S A 2012; 109 Suppl 2:17186-93. [PMID: 23045653 PMCID: PMC3477383 DOI: 10.1073/pnas.1121251109] [Citation(s) in RCA: 344] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The prefrontal cortex (PFC) receives input from all other cortical regions and functions to plan and direct motor, cognitive, affective, and social behavior across time. It has a prolonged development, which allows the acquisition of complex cognitive abilities through experience but makes it susceptible to factors that can lead to abnormal functioning, which is often manifested in neuropsychiatric disorders. When the PFC is exposed to different environmental events during development, such as sensory stimuli, stress, drugs, hormones, and social experiences (including both parental and peer interactions), the developing PFC may develop in different ways. The goal of the current review is to illustrate how the circuitry of the developing PFC can be sculpted by a wide range of pre- and postnatal factors. We begin with an overview of prefrontal functioning and development, and we conclude with a consideration of how early experiences influence prefrontal development and behavior.
Collapse
Affiliation(s)
- Bryan Kolb
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, Canada.
| | | | | | | | | | | |
Collapse
|
18
|
Ueda M, Sugiura C, Ohno K, Kakita A, Hori A, Ohama E, Vinters HV, Miyata H. Immunohistochemical expression of fibroblast growth factor-2 in developing human cerebrum and epilepsy-associated malformations of cortical development. Neuropathology 2011; 31:589-98. [DOI: 10.1111/j.1440-1789.2011.01205.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
19
|
Irmady K, Zechel S, Unsicker K. Fibroblast growth factor 2 regulates astrocyte differentiation in a region-specific manner in the hindbrain. Glia 2011; 59:708-19. [DOI: 10.1002/glia.21141] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2009] [Accepted: 12/15/2010] [Indexed: 01/04/2023]
|
20
|
Hausott B, Vallant N, Schlick B, Auer M, Nimmervoll B, Obermair GJ, Schwarzer C, Dai F, Brand-Saberi B, Klimaschewski L. Sprouty2 and -4 regulate axon outgrowth by hippocampal neurons. Hippocampus 2011; 22:434-41. [PMID: 21240919 DOI: 10.1002/hipo.20910] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2010] [Indexed: 11/06/2022]
Abstract
Sprouty proteins act as negative feedback inhibitors of fibroblast growth factor (FGF) signaling. FGFs belong to the neurotrophic factors and are involved in axonal growth during development and repair. We investigated the expression of Sprouty isoforms in hippocampal neurons as well as the regulation of Sprouty2 and -4 during development and their role in axon growth. Sprouty2 and -4 were located in the nucleus, the cytoplasm, in dendrites, and axons of hippocampal neurons concentrated in growth cones. During development in vivo and differentiation in vitro, expression of Sprouty2 and -4 was gradually downregulated in hippocampal neurons. Between 5 and 24 days in culture expression of both Sprouty isoforms was reduced by 70%. In vivo expression of Sprouty2 was reduced by 79% and of Sprouty4 by 93% on postnatal day 14 compared to embryonic day 16.5. Downregulation of Sprouty2 and -4 by shRNAs strongly promoted elongative axon growth by cultured hippocampal neurons, which was further increased by FGF-2 treatment. In addition, FGF-2 reduced expression of Sprouty2 by 33% and of Sprouty4 by 44%. Together, our results imply that Sprouty2 and -4 are downregulated in the hippocampus during postnatal brain development and that they can act as regulators of developmental axon growth.
Collapse
Affiliation(s)
- Barbara Hausott
- Division of Neuroanatomy, Innsbruck Medical University, Muellerstrasse 59, A-6020 Innsbruck, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Effect of Basic Fibroblast Growth Factor on Hippocampal Cholinergic Neurons in a Rodent Model of Ischaemic Encephalopathy. Basic Clin Pharmacol Toxicol 2010; 107:931-9. [DOI: 10.1111/j.1742-7843.2010.00603.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
22
|
Fahmy GH, Moftah MZ. Fgf-2 in astroglial cells during vertebrate spinal cord recovery. Front Cell Neurosci 2010; 4:129. [PMID: 21119776 PMCID: PMC2990542 DOI: 10.3389/fncel.2010.00129] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2010] [Accepted: 09/19/2010] [Indexed: 11/13/2022] Open
Abstract
Fibroblast growth factor-2 is a pleiotrophic cytokine with neurotrophic and gliogenic properties. It is known to regulate CNS injury responses, which include transformation of reactive astrocytes, neurogenesis, and promotion of neurotrophic activities. In the brain, it is localized in astrocytes and discrete neuronal populations. Following both central and peripheral nervous system injury, astrocytes become reactive. These activated cells undergo hypertrophy. A key indicator of astrocyte activation is the increased accumulation of intermediate filaments composed of glial fibrillary acidic protein (GFAP). Following physical insult of brain or spinal cord, reactive astrocytes show increased FGF-2 immunoreactivity. Thus, FGF-2 appears to participate in astrocytic differentiation and proliferation and a good candidate for astrocytic function regulation in healthy, injured, or diseased CNS. To further investigate the cellular mechanisms underlying FGF-2 restorative actions and to analyze the changes within astroglial cells, we studied the localization of GFAP and FGF-2 in adult intact and injured Pleurodeles CNS. Our results show that spinal cord injury triggers a significant increase in FGF-2 immunoreactivity in reactive astrocytes at sites of insult. In addition, these results were time-dependent. Increase in FGF-2 immunoreactivity along the CNS axis, starting 1-week post-injury, was long-lasting extending to 6 weeks. This increase was accompanied by an increase in GFAP immunoreactivity in the same spatial pattern except in SC3 where its level was almost similar to sham-operated animals. Therefore, we suggest that FGF-2 may be involved in cell proliferation and/or astroglial cells differentiation after body spinal cord transection, and could thus play an important role in locomotion recovery.
Collapse
Affiliation(s)
- Gehan H Fahmy
- Zoology Department, Faculty of Science, Alexandria University Alexandria, Egypt
| | | |
Collapse
|
23
|
Zechel S, Werner S, Unsicker K, von Bohlen und Halbach O. Expression and Functions of Fibroblast Growth Factor 2 (FGF-2) in Hippocampal Formation. Neuroscientist 2010; 16:357-73. [DOI: 10.1177/1073858410371513] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Among the 23 members of the fibroblast growth factor (FGF) family, FGF-2 is the most abundant one in the central nervous system. Its impact on neural cells has been profoundly investigated by in vitro and in vivo studies as well as by gene knockout analyses during the past 2 decades. Key functions of FGF-2 in the nervous system include roles in neurogenesis, promotion of axonal growth, differentiation in development, and maintenance and plasticity in adulthood. From a clinical perspective, its prominent role for the maintenance of lesioned neurons (e.g., ischemia and following transection of fiber tracts) is of particular relevance. In the unlesioned brain, FGF-2 is involved in synaptic plasticity and processes attributed to learning and memory. The focus of this review is on the expression of FGF-2 and its receptors in the hippocampal formation and the physiological and pathophysiological roles of FGF-2 in this region during development and adulthood.
Collapse
Affiliation(s)
- Sabrina Zechel
- Division of Molecular Neurobiology, Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Sandra Werner
- Department of Molecular Embryology, Institute of Anatomy & Cell Biology, University of Freiburg, Freiburg, Germany
| | - Klaus Unsicker
- Department of Molecular Embryology, Institute of Anatomy & Cell Biology, University of Freiburg, Freiburg, Germany
| | | |
Collapse
|
24
|
Yagami T, Takase K, Yamamoto Y, Ueda K, Takasu N, Okamura N, Sakaeda T, Fujimoto M. Fibroblast growth factor 2 induces apoptosis in the early primary culture of rat cortical neurons. Exp Cell Res 2010; 316:2278-90. [PMID: 20381486 DOI: 10.1016/j.yexcr.2010.03.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2009] [Revised: 03/24/2010] [Accepted: 03/27/2010] [Indexed: 11/16/2022]
Abstract
In the central nervous system, fibroblast growth factor 2 (FGF2) is known to have important functions in cell survival and differentiation. In addition to its roles as a neurotrophic factor, we found that FGF2 caused cell death in the early primary culture of cortical neurons. FGF2-induced neuronal cell death showed apoptotic characters, e.g., chromatin condensation and DNA fragmentation. The ultrastructural morphology of FGF2-treated neurons indicated apoptotic features such as progressive cell shrinkage, blebbing of the plasma membrane, loss of cytosolic organelles, clumping of chromatin, and fragmentation of DNA. Tyrosine kinase inhibitors significantly rescued neurons from FGF2-induced apoptosis. FGF2 potentiated a marked influx of Ca(2+) into neurons before apoptosis. Both a calcium chelator and L-type voltage-sensitive Ca(2+) channel (L-VSCC) blockers attenuated FGF2-induced apoptosis, whereas other blockers of VSCCs such as N-type and P/Q-types did not. Blockers of L-VSCCs significantly suppressed FGF2-enhanced Ca(2+) influx into neurons. Moreover, FGF2 also generated reactive oxygen species (ROS) before apoptosis. Radical scavengers reduced not only the FGF2-generated ROS, but also the FGF2-induced Ca(2+) influx and apoptosis. In conclusion, we demonstrated that FGF2 caused apoptosis via L-VSCCs in the early neuronal culture.
Collapse
Affiliation(s)
- Tatsurou Yagami
- Division of Physiology, Department of Pharmaceutical Health Care, Faculty of Pharmaceutical Sciences, Himeji Dokkyo University, 2-1, kami-ohno 7-Chome, Himeji, Hyogo, 670-8524, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Annenkov A. The insulin-like growth factor (IGF) receptor type 1 (IGF1R) as an essential component of the signalling network regulating neurogenesis. Mol Neurobiol 2009; 40:195-215. [PMID: 19714501 DOI: 10.1007/s12035-009-8081-0] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2009] [Accepted: 08/14/2009] [Indexed: 02/07/2023]
Abstract
The insulin-like growth factor receptor type 1 (IGF1R) signalling pathway is activated in the mammalian nervous system from early developmental stages. Its major effect on developing neural cells is to promote their growth and survival. This pathway can integrate its action with signalling pathways of growth and morphogenetic factors that induce cell fate specification and selective expansion of specified neural cell subsets. This suggests that during developmental and adult neurogenesis cellular responses to many signalling factors, including ligands of Notch, sonic hedgehog, fibroblast growth factor family members, ligands of the epidermal growth factor receptor, bone morphogenetic proteins and Wingless and Int-1, may be modified by co-activation of the IGF1R. Modulation of cell migration is another possible role that IGF1R activation may play in neurogenesis. Here, I briefly overview neurogenesis and discuss a role for IGF1R-mediated signalling in the developing and mature nervous system with emphasis on crosstalk between the signalling pathways of the IGF1R and other factors regulating neural cell development and migration. Studies on neural as well as on non-neural cells are highlighted because it may be interesting to test in neurogenic paradigms some of the models based on the information obtained in studies on non-neural cell types.
Collapse
Affiliation(s)
- Alexander Annenkov
- William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, UK.
| |
Collapse
|
26
|
Lin G, Goldman JE. An FGF-responsive astrocyte precursor isolated from the neonatal forebrain. Glia 2009; 57:592-603. [PMID: 19031440 DOI: 10.1002/glia.20788] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Gliogenesis in the mammalian CNS continues after birth, with astrocytes being generated well into the first two postnatal weeks. In this study, we have isolated an A2B5(+) astrocyte precursor (APC) from the postnatal rat forebrain, which is capable of differentiating into mature astrocytes in serum-free medium without further trophic support. Exposure to basic fibroblast growth factor (bFGF) selectively induces the APCs to proliferate, forming clusters of vimentin(+) cells, which, within 2 weeks, differentiate into GFAP(+) astrocytes. While bFGF functions as a potent mitogen, neither is it necessary to induce or maintain astrocyte differentiation, nor is it capable of maintaining the precursors in an immature, proliferative state. APCs exit the cell cycle and differentiate, even in the continued presence of fibroblast growth factor alone or in combination with other mitogenic factors such as platelet-derived growth factor. Under the culture conditions used, it was not possible to cause the astrocytes to re-enter cell cycle. After transplantation into the neonatal forebrain, APCs differentiated exclusively into astrocytes, regardless of brain region. Initially distributed widely within the forebrain, the precursors are most greatly concentrated within the subventricular zone (SVZ) and subcortical white matter, where they are maintained throughout postnatal development. APCs can be isolated from the SVZ and white matter of animals as late as 4 weeks after birth.
Collapse
Affiliation(s)
- Grace Lin
- Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | | |
Collapse
|
27
|
Zechel S, Unsicker K, von Bohlen und Halbach O. Fibroblast growth factor-2 deficiency affects hippocampal spine morphology, but not hippocampal catecholaminergic or cholinergic innervation. Dev Dyn 2009; 238:343-50. [DOI: 10.1002/dvdy.21839] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
28
|
Hossain WA, D'Sa C, Morest DK. Interactive roles of fibroblast growth factor 2 and neurotrophin 3 in the sequence of migration, process outgrowth, and axonal differentiation of mouse cochlear ganglion cells. J Neurosci Res 2008; 86:2376-91. [PMID: 18438927 DOI: 10.1002/jnr.21685] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
A growth factor may have different actions depending on developmental stage. We investigated this phenomenon in the interactions of fibroblast growth factor 2 (FGF2) and neurotrophins on cochlear ganglion (CG) development. The portions of the otocyst fated to form the CG and cochlear epithelium were cocultured at embryonic day 11 (E11). Cultures were divided into groups fed with defined medium, with or without FGF2 and neurotrophin supplements, alone or in combination, for 7 days. We measured the number of migrating neuroblasts and distances migrated, neurite outgrowth, and axonlike processes. We used immunohistochemistry to locate neurotrophin 3 (NT3) and its high-affinity receptor (TrkC) in the auditory system, along with FGF2 and its R1 receptor, at comparable developmental stages in vitro and in situ from E11 until birth (P1) in the precursors of hair cells, support cells, and CG cells. Potential sites for interaction were localized to the nucleus, perikaryal cytoplasm, and cell surfaces, including processes and growth cones. Time-lapse imaging and quantitative measures support the hypothesis that FGF2 alone or combined with neurotrophins promotes migration and neurite outgrowth. Synergism or antagonism between NT3 and other factors suggest interactions at the receptor level. Formation of axons, endings, and synaptic vesicle protein 2 were increased by interactions of NT3 and FGF2. Similar experiments with a mutant overexpressor for FGF2 suggest that endogenous FGF2 supports migration and neurite outgrowth of CG neuroblasts as well as proliferation, leading to accelerated development. The findings suggest interactive and sequential roles for FGF2 and NT3.
Collapse
Affiliation(s)
- Waheeda A Hossain
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut 06030-3401, USA
| | | | | |
Collapse
|
29
|
Audigier S, Guiramand J, Prado-Lourenco L, Conte C, Gonzalez-Herrera IG, Cohen-Solal C, Récasens M, Prats AC. Potent activation of FGF-2 IRES-dependent mechanism of translation during brain development. RNA (NEW YORK, N.Y.) 2008; 14:1852-64. [PMID: 18676616 PMCID: PMC2525950 DOI: 10.1261/rna.790608] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Fibroblast growth factor-2 (FGF-2) plays a fundamental role in brain functions. This role may be partly achieved through the control of its expression at the translational level via an internal ribosome entry site (IRES)-dependent mechanism. Transgenic mice expressing a bicistronic mRNA allowed us to study in vivo and ex vivo where this translational mechanism operates. Along brain development, we identified a stringent spatiotemporal regulation of FGF-2 IRES activity showing a peak at post-natal day 7 in most brain regions, which is concomitant with neuronal maturation. At adult age, this activity remained relatively high in forebrain regions. By the enrichment of this activity in forebrain synaptoneurosomes and by the use of primary cultures of cortical neurons or cocultures with astrocytes, we showed that this activity is indeed localized in neurons, is dependent on their maturation, and correlates with endogenous FGF-2 protein expression. In addition, this activity was regulated by astrocyte factors, including FGF-2, and spontaneous electrical activity. Thus, neuronal IRES-driven translation of the FGF-2 mRNA may be involved in synapse formation and maturation.
Collapse
Affiliation(s)
- Sylvie Audigier
- Institut National de la Santé et de la Recherche Médicale (INSERM), U858, Toulouse, France.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Sugiura C, Miyata H, Ueda M, Ohama E, Vinters HV, Ohno K. Immunohistochemical expression of fibroblast growth factor (FGF)-2 in epilepsy-associated malformations of cortical development (MCDs). Neuropathology 2008; 28:372-81. [DOI: 10.1111/j.1440-1789.2007.00881.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
31
|
Schmidt HD, Duman RS. The role of neurotrophic factors in adult hippocampal neurogenesis, antidepressant treatments and animal models of depressive-like behavior. Behav Pharmacol 2007; 18:391-418. [PMID: 17762509 DOI: 10.1097/fbp.0b013e3282ee2aa8] [Citation(s) in RCA: 498] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Major depressive disorder (MDD) is characterized by structural and neurochemical changes in limbic structures, including the hippocampus, that regulate mood and cognitive functions. Hippocampal atrophy is observed in patients with depression and this effect is blocked or reversed by antidepressant treatments. Brain-derived neurotrophic factor and other neurotrophic/growth factors are decreased in postmortem hippocampal tissue from suicide victims, which suggests that altered trophic support could contribute to the pathophysiology of MDD. Preclinical studies demonstrate that exposure to stress leads to atrophy and cell loss in the hippocampus as well as decreased expression of neurotrophic/growth factors, and that antidepressant administration reverses or blocks the effects of stress. Accumulating evidence suggests that altered neurogenesis in the adult hippocampus mediates the action of antidepressants. Chronic antidepressant administration upregulates neurogenesis in the adult hippocampus and this cellular response is required for the effects of antidepressants in certain animal models of depression. Here, we review cellular (e.g. adult neurogenesis) and behavioral studies that support the neurotrophic/neurogenic hypothesis of depression and antidepressant action. Aberrant regulation of neuronal plasticity, including neurogenesis, in the hippocampus and other limbic nuclei may result in maladaptive changes in neural networks that underlie the pathophysiology of MDD.
Collapse
Affiliation(s)
- Heath D Schmidt
- Division of Molecular Psychiatry, Abraham Ribicoff Research Facilities, Department of Psychiatry and Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA
| | | |
Collapse
|
32
|
Slotkin TA, Seidler FJ, Fumagalli F. Exposure to organophosphates reduces the expression of neurotrophic factors in neonatal rat brain regions: similarities and differences in the effects of chlorpyrifos and diazinon on the fibroblast growth factor superfamily. ENVIRONMENTAL HEALTH PERSPECTIVES 2007; 115:909-16. [PMID: 17589599 PMCID: PMC1892141 DOI: 10.1289/ehp.9901] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2006] [Accepted: 02/27/2007] [Indexed: 05/16/2023]
Abstract
BACKGROUND The fibroblast growth factor (FGF) superfamily of neurotrophic factors plays critical roles in neural cell development, brain assembly, and recovery from neuronal injury. OBJECTIVES We administered two organophosphate pesticides, chlorpyrifos and diazinon, to neonatal rats on postnatal days 1-4, using doses below the threshold for systemic toxicity or growth impairment, and spanning the threshold for barely detectable cholinesterase inhibition: 1 mg/kg/day chlorpyrifos and 1 or 2 mg/kg/day diazinon. METHODS Using microarrays, we then examined the regional expression of mRNAs encoding the FGFs and their receptors (FGFRs) in the forebrain and brain stem. RESULTS Chlorpyrifos and diazinon both markedly suppressed fgf20 expression in the forebrain and fgf2 in the brain stem, while elevating brain stem fgfr4 and evoking a small deficit in brain stem fgf22. However, they differed in that the effects on fgf2 and fgfr4 were significantly larger for diazinon, and the two agents also showed dissimilar, smaller effects on fgf11, fgf14, and fgfr1. CONCLUSIONS The fact that there are similarities but also notable disparities in the responses to chlorpyrifos and diazinon, and that robust effects were seen even at doses that do not inhibit cholinesterase, supports the idea that organophosphates differ in their propensity to elicit developmental neurotoxicity, unrelated to their anticholinesterase activity. Effects on neurotrophic factors provide a mechanistic link between organophosphate injury to developing neurons and the eventual, adverse neurodevelopmental outcomes.
Collapse
Affiliation(s)
- Theodore A Slotkin
- Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, USA.
| | | | | |
Collapse
|
33
|
Chadashvili T, Peterson DA. Cytoarchitecture of fibroblast growth factor receptor 2 (FGFR-2) immunoreactivity in astrocytes of neurogenic and non-neurogenic regions of the young adult and aged rat brain. J Comp Neurol 2006; 498:1-15. [PMID: 16856175 DOI: 10.1002/cne.21009] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Fibroblast growth factors (FGFs) are polypeptides that exert diverse biological effects on many cell types and tissues during embryogenesis and adulthood. In the adult brain, FGF-2 is primarily expressed by astrocytes and select groups of neurons. It has been shown that FGF-2 is neuroprotective and can stimulate proliferation of NSCs in neurogenic regions of the adult mammalian brain. Cellular responses to FGFs are mediated through membrane-spanning tyrosine kinase receptors in conjunction with low affinity binding to heparin sulfate proteoglycans. Four FGF receptors (FGFR1-4) have been cloned and characterized to date. In this study, we describe the anatomical distribution of FGFR-2 in young and aged rat brains. We demonstrate that the olfactory bulb, hippocampus, and cerebellum display the most robust FGFR-2 expression and observed age-related decrease in FGFR-2 levels in some but not all brain regions. In addition, we identified astrocytes as the primary source of FGFR-2 expression using immunofluorescence confocal microscopy. The astrocyte populations in the neurogenic areas, the subventricular zone (SVZ) and the subgranular zone (SGZ) of the dentate gyrus, express high levels of FGFR-2 protein, which points to its possible involvement in neurogenesis. We also explored the role of FGFR-2 in response to perforant pathway lesion and observed enhanced FGFR-2 expression by astrocytes surrounding the lesion. Thus, FGF-2 biological effects on astrocytes appear to be mediated through FGFR-2-dependent mechanisms, and this may provide an indirect route by which FGF-2 acts on neuronal populations.
Collapse
Affiliation(s)
- Tamuna Chadashvili
- Neural Repair and Neurogenesis Laboratory, Department of Neuroscience, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, Chicago, Illinois 60064, USA
| | | |
Collapse
|
34
|
Monfils MH, Driscoll I, Melvin NR, Kolb B. Differential expression of basic fibroblast growth factor-2 in the developing rat brain. Neuroscience 2006; 141:213-21. [PMID: 16677765 DOI: 10.1016/j.neuroscience.2006.03.047] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2005] [Revised: 01/25/2006] [Accepted: 03/23/2006] [Indexed: 11/21/2022]
Abstract
Basic fibroblast growth factor-2 is a trophic molecule involved in a number of functions within the CNS, including the regulation of CNS responses to injury. Prior studies suggest that rats recover differently from injury inflicted to different regions and at different ages throughout development, and that basic fibroblast growth factor-2 may, at least in part, underlie this phenomenon. In the present study, we describe the distribution of basic fibroblast growth factor-2 at postnatal days 0, 2, 6, 10, 12, 14, 18, 21 and 30 in the indusium griseum, the external capsule, the hippocampus, the medial prefrontal cortex, the motor cortex, the rostral migratory stream, and the subventricular zone. Our results suggest a differential temporal and spatial expression of basic fibroblast growth factor-2 throughout development, which may explain the differential recovery observed from cortical lesions inflicted at different time points after birth.
Collapse
Affiliation(s)
- M-H Monfils
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, Alberta, Canada.
| | | | | | | |
Collapse
|
35
|
Hattiangady B, Rao MS, Zaman V, Shetty AK. Incorporation of embryonic CA3 cell grafts into the adult hippocampus at 4-months after injury: effects of combined neurotrophic supplementation and caspase inhibition. Neuroscience 2006; 139:1369-83. [PMID: 16580143 DOI: 10.1016/j.neuroscience.2006.01.058] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2005] [Revised: 01/17/2006] [Accepted: 01/26/2006] [Indexed: 10/24/2022]
Abstract
As receptivity of the injured hippocampus to cell grafts decreases with time after injury, strategies that improve graft integration are necessary for graft-mediated treatment of chronic neurodegenerative conditions such as temporal lobe epilepsy. We ascertained the efficacy of two distinct graft-augmentation strategies for improving the survival of embryonic day 19 hippocampal CA3 cell grafts placed into the adult hippocampus at 4-months after kainic acid induced injury. The donor cells were labeled with 5'-bromodeoxyuridine, and pre-treated and grafted with either brain-derived neurotrophic factor, neurotrophin-3 and a caspase inhibitor or fibroblast growth factor and caspase inhibitor. The yield of surviving grafted cells and neurons were quantified at 2-months post-grafting. The yield of surviving cells was substantially greater in grafts treated with brain-derived neurotrophic factor, neurotrophin-3 and caspase inhibitor (84%) or fibroblast growth factor and caspase inhibitor (99% of injected cells) than standard cell grafts (26%). Because approximately 85% of surviving grafted cells were neurons, increased yield in augmented groups reflects enhanced survival of grafted neurons. Evaluation of the mossy fiber synaptic re-organization in additional kainic acid-lesioned rats receiving grafts enriched with brain-derived neurotrophic factor, neurotrophin-3 and caspase inhibitor at 3-months post-grafting revealed reduced aberrant dentate mossy fiber sprouting in the dentate supragranular layer than "lesion-only" rats at 4 months post-kainic acid, suggesting that some of the aberrantly sprouted mossy fibers in the dentate supragranular layer withdraw when apt target cells (i.e. grafted neurons) become available in their vicinity. Thus, the yield of surviving neurons from CA3 cell grafts placed into the adult hippocampus at an extended time-point after injury could be enhanced through apt neurotrophic supplementation and caspase inhibition. Apt grafting is also efficacious for reversing some of the abnormal synaptic reorganization prevalent in the hippocampus at later time-points after injury.
Collapse
Affiliation(s)
- B Hattiangady
- Department of Surgery (Neurosurgery), Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | |
Collapse
|
36
|
Rao MS, Hattiangady B, Shetty AK. Fetal hippocampal CA3 cell grafts enriched with FGF-2 and BDNF exhibit robust long-term survival and integration and suppress aberrant mossy fiber sprouting in the injured middle-aged hippocampus. Neurobiol Dis 2005; 21:276-90. [PMID: 16099669 DOI: 10.1016/j.nbd.2005.07.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2005] [Revised: 06/20/2005] [Accepted: 07/06/2005] [Indexed: 11/27/2022] Open
Abstract
Cell transplants that successfully replace the lost neurons and facilitate the reconstruction of the disrupted circuitry in the injured aging hippocampus are invaluable for treating acute head injury, stroke and status epilepticus in the elderly. This is because apt graft integration has the potential to prevent the progression of the acute injury into chronic epilepsy in the elderly. However, neural transplants into the injured middle-aged or aged hippocampus exhibit poor cell survival, suggesting that apt graft augmentation strategies are critical for robust integration of grafted cells into the injured aging hippocampus. We examined the efficacy of pre-treatment and grafting of donor fetal CA3 cells with a blend of fibroblast growth factor-2 (FGF-2) and brain-derived neurotrophic factor (BDNF) for lasting survival and integration of grafted cells in the injured middle-aged (12 months old) hippocampus of F344 rats. Grafts were placed at 4 days after the kainic-acid-induced hippocampal injury and were analyzed at 6 months post-grafting. We demonstrate that 80% of grafted cells exhibit prolonged survival and 71% of grafted cells differentiate into CA3 pyramidal neurons. Grafts also receive a robust afferent input from the host mossy fibers and project efferent axons into the denervated zones of the dentate gyrus and the CA1 subfield. Consequently, the aberrant sprouting of the dentate mossy fibers, an epileptogenic change that typically ensues after the hippocampal injury, was suppressed. Thus, grafts of fetal CA3 cells enriched with FGF-2 and BDNF exhibit robust integration and dampen the abnormal mossy fiber sprouting in the injured middle-aged hippocampus. Because the aberrantly sprouted mossy fibers contribute to the generation of seizures, the results suggest that the grafting intervention using FGF-2 and BDNF is efficacious for suppressing epileptogenesis in the injured middle-aged hippocampus.
Collapse
Affiliation(s)
- Muddanna S Rao
- Medical Research and Surgery Services, Veterans Affairs Medical Center, Durham NC 27705, USA
| | | | | |
Collapse
|
37
|
Pavlov I, Lauri S, Taira T, Rauvala H. The role of ECM molecules in activity-dependent synaptic development and plasticity. ACTA ACUST UNITED AC 2004; 72:12-24. [PMID: 15054901 DOI: 10.1002/bdrc.20001] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Growth and guidance of neurites (axons and dendrites) during development is the prerequisite for the establishment of functional neural networks in the adult organism. In the adult, mechanisms similar to those used during development may regulate plastic changes that underlie important nervous system functions, such as memory and learning. There is now ever-increasing evidence that extracellular matrix (ECM)-associated factors are critically involved in the formation of neuronal connections during development, and their plastic changes in the adult. Here, we review the current literature on the role of ECM components in activity-dependent synaptic development and plasticity, with the major focus on the thrombospondin type I repeat (TSR) domain-containing proteins. We propose that ECM components may modulate neuronal development and plasticity by: 1) regulating cellular motility and morphology, thus contributing to structural alterations that are associated with the expression of synaptic plasticity, 2) coordinating transsynaptic signaling during plasticity via their cell surface receptors, and 3) defining the physical parameters of the extracellular space, thereby regulating diffusion of soluble signaling molecules in the extracellular space (ECS).
Collapse
Affiliation(s)
- Ivan Pavlov
- Neuroscience Center and Department of Biosciences, University of Helsinki, Helsinki, Finland
| | | | | | | |
Collapse
|
38
|
Functions of fibroblast growth factor (FGF)-2 and FGF-5 in astroglial differentiation and blood-brain barrier permeability: evidence from mouse mutants. J Neurosci 2003. [PMID: 12878680 DOI: 10.1523/jneurosci.23-16-06404.2003] [Citation(s) in RCA: 105] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Multiple evidence suggests that fibroblast growth factors (FGFs), most prominently FGF-2, affect astroglial proliferation, maturation, and transition to a reactive phenotype in vitro, and after exogenous administration, in vivo. Whether this reflects a physiological role of endogenous FGF is unknown. Using FGF-2 and FGF-5 single- and double mutant mice we show now a region-specific reduction of glial fibrillary acidic protein (GFAP), but not of S100 in gray matter astrocytes. FGF-2 is apparently the major regulator of GFAP, because in mice deficient for FGF-2, GFAP is distinctly reduced in cortex and striatum, whereas in FGF-5-/- animals only a reduction in the midbrain tegmentum can be observed. In FGF-2-/-/FGF-5-/- double mutant animals, GFAP-immunoreactivity is reduced in all three brain regions. Cortical astrocytes cultured from FGF-2-/-/FGF-5-/- double mutant mice revealed reduced levels of GFAP, but not S100 as compared with wild-type littermates. This phenotype could be rescued by exogenous FGF-2 but not FGF-5 (10 ng/ml). Electron microscopy revealed reduced levels of intermediate filaments in perivascular astroglial endfeet. This defect was accompanied by enhanced permeability of the blood-brain barrier (BBB), as detected by albumin extravasation. Levels of the tight junction proteins Occludin and ZO-1 were reduced in blood vessels of FGF-2-/-/FGF-5-/- double mutant mice as compared with wild-type littermates. Our data support the notion that endogenous FGF-2 and FGF-5 regulate GFAP expression in a region-specific manner. The observed defect in astroglial differentiation is accompanied by a defect in BBB function arguing for an indirect or direct role of FGFs in the regulation of BBB permeability in vivo.
Collapse
|
39
|
Zaman V, Shetty AK. Pretreatment of donor cells with FGF-2 enhances survival of fetal hippocampal CA3 cell transplants in the chronically lesioned young adult hippocampus. Exp Neurol 2003; 183:11-24. [PMID: 12957484 DOI: 10.1016/s0014-4886(03)00167-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The lesioned CA3 region of the young adult hippocampus is very conducive for robust survival and integration of fetal hippocampal CA3 cell grafts when transplanted at an early postlesion delay of 4 days. However, similar CA3 cell grafts placed at 45 days postlesion display significantly diminished cell survival, implying that the receptivity of the lesioned young adult host hippocampus to grafts decreases considerably with a prolonged postlesion transplantation delay. We hypothesize that decreased cell survival in grafts placed into the chronically lesioned hippocampus is due to a reduced level of host neurotrophic factors that support fetal hippocampal cells; hence, pretreatment and grafting of donor fetal CA3 cells with fibroblast growth factor-2 (FGF-2) considerably enhances graft neuronal integration into the chronically lesioned young adult hippocampus. We employed the optical fractionator cell counting method and rigorously quantified the number of surviving cells and neurons derived from 5'-bromodeoxyuridine-labeled Embryonic Day 19 CA3 cell grafts pretreated and transplanted with FGF-2 into the lesioned CA3 region of the young adult rat hippocampus, at a delay of 60 days after a unilateral intracerebroventricular administration of the kainic acid. For comparison, we also analyzed the survival of standard fetal CA3 cell grafts (i.e., without FGF-2 treatment) after similar transplantation. Pre treatment and transplantation of CA3 cell grafts with FGF-2 resulted in a robust yield of surviving cells (115% of injected cells) and neurons (100% of injected cells) from grafts. In contrast, standard CA3 cell grafts exhibited a reduced yield of surviving cells (29%) and neurons (25%). Thus, the yield of neurons from fetal hippocampal CA3 cell grafts placed into the chronically lesioned young adult hippocampus can be greatly enhanced by a simple pretreatment and grafting of donor fetal CA3 cells with FGF-2. These results have significance toward advancement of clinically feasible cell grafting strategies for repair of the damaged young adult hippocampus, particularly at extended periods after the injury or the onset of neurodegenerative diseases.
Collapse
Affiliation(s)
- Vandana Zaman
- Medical Research Service, Veterans Affairs Medical Center, Durham, NC 27705, USA
| | | |
Collapse
|
40
|
Morale MC, Gallo F, Tirolo C, L'Episcopo F, Gennuso F, Testa N, Caniglia S, Spina-Purrello V, Avola R, Scoto GM, Marchetti B. The reproductive system at the neuroendocrine-immune interface: focus on LHRH, estrogens and growth factors in LHRH neuron-glial interactions. Domest Anim Endocrinol 2003; 25:21-46. [PMID: 12963097 DOI: 10.1016/s0739-7240(03)00043-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Bidirectional communication between the neuroendocrine and immune systems plays a pivotal role in health and disease. Signals generated by the hypothalamic-pituitary-gonadal (HPG) axis (i.e. luteinizing hormone-releasing hormone, LHRH, and sex steroids) are major players coordinating the development immune system function. Conversely, products generated by immune system activation exert powerful and longlasting effects on HPG axis activity. In the central nervous system (CNS), one chief neuroendocrine-immune (NEI) compartment is represented by the astroglial cell population and its mediators. Of special interest, the major supporting cells of the brain and the thymus, astrocytes and thymic epithelial cells, share a similar origin and a similar set of peptides, transmitters, hormones and cytokines functioning as paracrine/autocrine regulators. This may explain some fundamental analogies in LHRH regulation of both cell types during ontogeny and in adult life. Hence, the neuropeptide LHRH significantly modulates astrocyte and thymic cell development and function. Here we focus this work on LHRH neuron-glial signaling cascades which dictate major changes during LHRH neuronal differentiation and growth as well as in response to hormonal manipulations and pro-inflammatory challenges. The interplay between LHRH, growth factors, estrogens and pro-inflammatory mediators will be discussed, and the potential physiopathological implications of these findings summarized. The overall study highlights the plasticity of this intersystem cross-talk and emphasize neuron-glial interactions as a key regulatory level of neuroendocrine axes activity.
Collapse
Affiliation(s)
- M C Morale
- Department of Neuropharmacology, OASI Institute for Research and Care (IRCCS) on Mental Retardation and Brain Aging, Via Conte Ruggero 73, 94018 Troina (EN), Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Pringle NP, Yu WP, Howell M, Colvin JS, Ornitz DM, Richardson WD. Fgfr3 expression by astrocytes and their precursors: evidence that astrocytes and oligodendrocytes originate in distinct neuroepithelial domains. Development 2003; 130:93-102. [PMID: 12441294 DOI: 10.1242/dev.00184] [Citation(s) in RCA: 121] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The postnatal central nervous system (CNS) contains many scattered cells that express fibroblast growth factor receptor 3 transcripts (Fgfr3). They first appear in the ventricular zone (VZ) of the embryonic spinal cord in mid-gestation and then distribute into both grey and white matter - suggesting that they are glial cells, not neurones. The Fgfr3(+) cells are interspersed with but distinct from platelet-derived growth factor receptor alpha (Pdgfra)-positive oligodendrocyte progenitors. This fits with the observation that Fgfr3 expression is preferentially excluded from the pMN domain of the ventral VZ where Pdgfra(+) oligodendrocyte progenitors--and motoneurones--originate. Many glial fibrillary acidic protein (Gfap)- positive astrocytes co-express Fgfr3 in vitro and in vivo. Fgfr3(+) cells within and outside the VZ also express the astroglial marker glutamine synthetase (Glns). We conclude that (1) Fgfr3 marks astrocytes and their neuroepithelial precursors in the developing CNS and (2) astrocytes and oligodendrocytes originate in complementary domains of the VZ. Production of astrocytes from cultured neuroepithelial cells is hedgehog independent, whereas oligodendrocyte development requires hedgehog signalling, adding further support to the idea that astrocytes and oligodendrocytes can develop independently. In addition, we found that mice with a targeted deletion in the Fgfr3 locus strongly upregulate Gfap in grey matter (protoplasmic) astrocytes, implying that signalling through Fgfr3 normally represses Gfap expression in vivo.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Astrocytes/cytology
- Astrocytes/physiology
- Cells, Cultured
- Central Nervous System/cytology
- Central Nervous System/embryology
- Chick Embryo
- Epithelium/embryology
- Gene Expression Regulation, Developmental
- Glial Fibrillary Acidic Protein/genetics
- Glial Fibrillary Acidic Protein/metabolism
- Glutamate-Ammonia Ligase/genetics
- Glutamate-Ammonia Ligase/metabolism
- Hedgehog Proteins
- Mice
- Mice, Inbred C57BL
- Mice, Mutant Strains
- Oligodendroglia/cytology
- Oligodendroglia/physiology
- Protein-Tyrosine Kinases
- Rats
- Rats, Sprague-Dawley
- Receptor, Fibroblast Growth Factor, Type 3
- Receptor, Platelet-Derived Growth Factor alpha/genetics
- Receptor, Platelet-Derived Growth Factor alpha/metabolism
- Receptors, Fibroblast Growth Factor/genetics
- Receptors, Fibroblast Growth Factor/metabolism
- Spinal Cord/cytology
- Spinal Cord/embryology
- Spinal Cord/metabolism
- Stem Cells/cytology
- Stem Cells/physiology
- Trans-Activators/genetics
- Trans-Activators/metabolism
Collapse
Affiliation(s)
- Nigel P Pringle
- Wolfson Institute for Biomedical Research and Department of Biology, University College London, Gower Street, London WC1E 6BT, UK
| | | | | | | | | | | |
Collapse
|
42
|
Li AJ, Suzuki S, Suzuki M, Mizukoshi E, Imamura T. Fibroblast growth factor-2 increases functional excitatory synapses on hippocampal neurons. Eur J Neurosci 2002; 16:1313-24. [PMID: 12405992 DOI: 10.1046/j.1460-9568.2002.02193.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The effect of fibroblast growth factor-2 (FGF-2) on synapse formation was investigated using rat cultured hippocampal neurons. Treatment with FGF-2 (0.4-10 ng/mL) for 6 days enhanced synaptogenesis on these neurons by approximately 50%, as determined by counting puncta immunostained for presynaptic- or postsynaptic-specific proteins. This enhancement was statistically significant, and was abolished by a specific inhibitor of mitogen-activated protein kinase (MAPK). The majority of neurons expressed FGF receptors (types 1-3) abundantly on the membrane of somata, dendrites, and growth cones, and in these regions phosphorylation of MAPK was enhanced after FGF-2 application. Furthermore, our experiments showed that the majority of synapses formed in cultures containing FGF-2 were positive both for presynaptic proteins and postsynaptic excitatory synapse-specific proteins, and that these synapses had a similar capacity to recycle the fluorescent styryl dye FM4-64 as those in the control culture. These results indicate that: (i) FGF-2 increases excitatory synapses on hippocampal neurons by activating MAPK activity through FGF receptors; and (ii) synapses formed in FGF-2-treated culture are capable of cycling vesicles.
Collapse
Affiliation(s)
- Ai-Jun Li
- Age Dimension Research Centre, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1 Higashi, Tsukuba, Ibaraki 305-8566, Japan
| | | | | | | | | |
Collapse
|
43
|
Winkler S, Stahl RC, Carey DJ, Bansal R. Syndecan-3 and perlecan are differentially expressed by progenitors and mature oligodendrocytes and accumulate in the extracellular matrix. J Neurosci Res 2002; 69:477-87. [PMID: 12210841 DOI: 10.1002/jnr.10311] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Oligodendrocyte progenitors originate in the subventricular zone, proliferate, migrate to their final destinations, differentiate, and interact with axons to produce multilamellar myelin sheaths. These processes are regulated by a variety of environmental signals, including growth factors, the extracellular matrix, and adhesion molecules. Heparan sulfate proteoglycans are premier candidates as participants in this regulation by virtue of their structural diversity and their capacity to function as coreceptors for both growth factors and extracellular matrix molecules. Consistently with this, we have previously shown that oligodendrocyte progenitors are unable to proliferate in response to fibroblast growth factor-2 (FGF-2) in the absence of sulfated heparan sulfate proteoglycan. Here we show that members of three families of heparan sulfate proteoglycans, syndecan, perlecan, and glypican, are developmentally and posttranscriptionally regulated during oligodendrocyte-lineage progression: Syndecan-3 is synthesized by oligodendrocyte progenitors (but not terminally differentiated oligodendrocytes) and is up-regulated by FGF-2; perlecan synthesis increases as oligodendrocytes undergo terminal differentiation; glypican-1 is expressed by both progenitors and differentiated oligodendrocytes. Astrocytes express glypican-1 and perlecan but not syndecan-3. All three of these heparan sulfate proteoglycans are shed from the cell surface and bind to specific substrates. The developmentally regulated expression of these heparan sulfate proteoglycans is indicative of their participation in events involving growth factor receptors and the extracellular matrix that may regulate oligodendrocyte progenitor proliferation, migration, and adhesion phenomena.
Collapse
Affiliation(s)
- Susan Winkler
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington 06030-3401, USA
| | | | | | | |
Collapse
|
44
|
Cheng Y, Black IB, DiCicco-Bloom E. Hippocampal granule neuron production and population size are regulated by levels of bFGF. Eur J Neurosci 2002; 15:3-12. [PMID: 11860501 DOI: 10.1046/j.0953-816x.2001.01832.x] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Numerous studies of the proliferative effects of basic fibroblast growth factor (bFGF) in culture, including neonatal and adult hippocampal precursors, suggest that the factor plays a ubiquitous and life-long role in neurogenesis. In contrast, in vivo, bFGF is devoid of effects on neurons in mature hippocampus, raising the possibility that bFGF exhibits developmental stage-specific activity in the complex animal environment. To define neurogenetic effects in the newborn, a single subcutaneous injection of bFGF (20 ng/gm) was administered to postnatal day 1 (P1) rats, and hippocampal DNA content was quantified: bFGF elicited an increase in total DNA throughout adulthood, by 48% at P4, 25% at P22, and 17% at P180, suggesting that bFGF increases hippocampal cell number. To define mechanisms, bromodeoxyuridine (BrdU) was injected at P1 and mitotically labelled cells were assessed at P22: there was a twofold increase in BrdU-positive cells in the dentate granule cell layer (GCL), indicating that bFGF enhanced the generation of neurons, or neuronogenesis, from a cohort of precursors. Moreover, enhanced mitosis and survival led to a 33% increase in absolute GCL neuron number, suggesting that neuron production depends on environmental levels of bFGF. To evaluate this possibility, bFGF-knockout mice were analyzed: hippocampal DNA content was decreased at all ages examined (P3, -42%; P21, -28%; P360, -18%), and total GCL neuron and glial fibrillary acidic protein (GFAP)-positive cell number were decreased by 30%, indicating that bFGF is necessary for normal hippocampal neurogenesis. We conclude that environmental levels of bFGF regulate neonatal hippocampal neurogenesis. As adult hippocampal neuronogenesis was unresponsive to bFGF manipulation in our previous study [Wagner, J.P., Black, I.B. & DiCicco-Bloom, E. (1999) J. Neurosci., 19, 6006], these observations suggest distinct, stage-specific roles of bFGF in the dentate gyrus granule cell lineage.
Collapse
Affiliation(s)
- Yinghong Cheng
- Department of Neuroscience and Cell Biology UMDNJ/Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | | | | |
Collapse
|
45
|
Zhao X, Lein ES, He A, Smith SC, Aston C, Gage FH. Transcriptional profiling reveals strict boundaries between hippocampal subregions. J Comp Neurol 2001; 441:187-96. [PMID: 11745644 DOI: 10.1002/cne.1406] [Citation(s) in RCA: 154] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The hippocampus consists of distinct anatomic regions that have been demonstrated to have different biological functions. To explore the molecular differences between hippocampal subregions, we performed transcriptional profiling analysis by using DNA microarray technology. The cRNA derived from the CA1, CA3, and dentate gyrus regions of the hippocampus and from spinal cord was hybridized to Affymetrix high-density oligo arrays. This systematic approach revealed sets of genes that were expressed specifically in subregions of the hippocampus corresponding to predefined cytoarchitectural boundaries, which could be confirmed by in situ hybridization and Real Time quantitative polymerase chain reaction. The relative enrichment and absence of genes in the hippocampal subregions support the conclusion that there is a molecular basis for the previously defined anatomic subregions of the hippocampus and also reveal genes that could be important in defining the unique functions of the hippocampal subfields.
Collapse
Affiliation(s)
- X Zhao
- Laboratory of Genetics, Vision Center Laboratory, the Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | | | | | | | | | | |
Collapse
|
46
|
Molteni R, Fumagalli F, Magnaghi V, Roceri M, Gennarelli M, Racagni G, Melcangi RC, Riva MA. Modulation of fibroblast growth factor-2 by stress and corticosteroids: from developmental events to adult brain plasticity. BRAIN RESEARCH. BRAIN RESEARCH REVIEWS 2001; 37:249-58. [PMID: 11744090 DOI: 10.1016/s0165-0173(01)00128-x] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Neurotrophic factors are a heterogeneous group of peptides that play important roles on brain function at different development stages. Basic fibroblast growth factor (FGF-2), one of these molecules, is highly expressed in developing and adult brain. Its expression can be regulated under different experimental situations and this may be relevant for cellular vulnerability and brain plasticity. Stress and glucocorticoid hormones produce short- and long-term effects on brain function, which can involve the regulation of specific neurotrophic factors within selected brain structures. Treatments with corticosterone or dexamethasone up-regulate FGF-2 expression in different rat brain regions as well as in cultured astroglial cells. A similar elevation of FGF-2 biosynthesis is also observed in several brain regions following an acute restraint stress. This response is rapid and transient and, as FGF-2 is neuroprotective, may represent a defense mechanism through which the brain may limit the deleterious effect of stress over time. Moreover exposure to corticosterone during late stage of embryonic life (E18-E20) produces a significant reduction of FGF-2 mRNA levels in the adult hippocampus of male rats as well as changes in its acute modulation in response to stress or corticosterone. These data suggest that stress-related events taking place during brain maturation can modulate the expression of FGF-2 within selected brain regions thus contributing to permanent structural and functional alterations leading to an increased vulnerability to challenging life events.
Collapse
Affiliation(s)
- R Molteni
- Center of Excellence for Neurodegenerative Disorders, University of Milan, Via Balzaretti 9, 20133, Milan, Italy
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Ford-Perriss M, Abud H, Murphy M. Fibroblast growth factors in the developing central nervous system. Clin Exp Pharmacol Physiol 2001; 28:493-503. [PMID: 11422214 DOI: 10.1046/j.1440-1681.2001.03477.x] [Citation(s) in RCA: 195] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
1. It is now clear that members of the fibroblast growth factor (FGF) family have multiple roles during the formation of the central nervous system (CNS). 2. There are at least 23 members of the FGF family and, of these, 10 are expressed in the developing CNS, along with four FGF receptors (FGFR-1-4). 3. The present review discusses the roles of these FGFs, with emphasis on FGF-2, FGF-8, FGF-15 and FGF-17. Fibroblast growth factors-2 and -15 are generally expressed throughout the developing CNS, whereas FGF-8 and FGF-17 are tightly localized to specific regions of the developing brain and are only expressed in the embryo during the early phases of proliferation and neurogenesis. 4. Expression studies on FGFRs in the chick and mouse indicate that FGFR-1 is most generally expressed, whereas FGFR-2 and FGFR-3 show highly localized but changing patterns of expression throughout CNS development. The FGFR-4 has been localized to the developing CNS in fish but not at a detailed level, as yet, in chick or mouse. 5. A picture is emerging from these studies that particular FGFs signal through specific receptors in a highly localized manner to regulate the development of different regions of the brain. 6. This picture has been demonstrated so far for the developing cortex (FGF-2-/- mice), the forebrain and midbrain (FGF-8 hypomorphs) and the cerebellum (FGF-17/FGF-8 mutant mice). In addition, generation of mutant animals deleted for FGFR-1 and FGFR-2b IIIb demonstrate their importance in FGF signalling. 7. However, there are significant gaps in our knowledge of the localization of members of the FGF family and their receptors. More detailed information on the spatio-temporal mapping of FGFs and FGFR isoforms is required in order to understand the molecular mechanisms through which FGFs signal.
Collapse
Affiliation(s)
- M Ford-Perriss
- Department of Anatomy and Cell Biology, The University of Melbourne, Victoria 3010, Australia.
| | | | | |
Collapse
|
48
|
Katsuki H, Itsukaichi Y, Matsuki N. Distinct signaling pathways involved in multiple effects of basic fibroblast growth factor on cultured rat hippocampal neurons. Brain Res 2000; 885:240-50. [PMID: 11102578 DOI: 10.1016/s0006-8993(00)02953-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
We investigated possible involvement of voltage-dependent Ca(2+) channels (VDCCs) and several intracellular signaling mechanisms in multiple actions of basic fibroblast growth factor (bFGF), such as survival promotion, induction of calbindin D(28k) expression as well as acceleration of neuritic branch formation of cultured rat hippocampal neurons. Immunocytochemical staining with anti-gamma-aminobutyric acid (GABA) antibody showed that the promotion of neuron survival by bFGF in high cell-density cultures were exerted exclusively on GABA-negative neurons. Nicardipine (5 microM) attenuated the effect of bFGF on neuronal survival and formation of neurite branches, suggesting that the activity of L-type VDCCs is required for these effects. In contrast, stimulation of calbindin expression by bFGF was not attenuated by nicardipine. A phospholipase C inhibitor U73122 (1 microM) prevented the effect of bFGF on neurite branch formation, but not on neuronal survival or calbindin expression. On the other hand, chronic application of phorbol-12-myristate-13-acetate (1 microM) inhibited the effect of bFGF on neuronal survival, without inhibiting the other bFGF actions. Forskolin (100 microM) attenuated the effect of bFGF on neuronal survival and neurite branch formation, indicating that cyclic AMP plays negative regulatory roles in these actions of bFGF. Taken together, these results suggest that multiple biological actions of bFGF on hippocampal neurons are exerted through, and modulated by, distinct signaling pathways.
Collapse
Affiliation(s)
- H Katsuki
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan.
| | | | | |
Collapse
|
49
|
Abstract
The development of place and cue spatial navigation was evaluated in 18-, 19-, and 20-day-old males in the Morris water task (MWT). Past work has suggested that place and cue learning develop at different rates, suggesting that the two aspects of spatial navigation have different neural substrates. In the present study, a new training methodology was used in which animals received spaced training trials, drying and warming in between trials to maintain body temperature, and two probe trial-dependent measures to evaluate spatial memory performance. All ages of rats had lower latencies on the cue task than on the place task. Nevertheless, 18-day-old rats did not learn either task as measured by acquisition latencies and probe trial-dependent measures. The 19- and 20-day-old rats learned both the place and cue tasks as measured by acquisition latency and direct swims to the correct platform location on the probe trial, and both 19- and 20-day-old rats demonstrated a strong spatial bias to the former platform location on the place probe trial but not on the cue probe trial. The finding that developmental onset of place and cue spatial navigation is rapid and complete by day 19 is discussed in relation to contemporary theories of spatial navigation.
Collapse
Affiliation(s)
- R W Brown
- Department of Psychology and Neuroscience, University of Lethbridge, Lethbridge, AB, Canada T1K 3M4
| | | |
Collapse
|
50
|
Gallo F, Morale MC, Tirolo C, Testa N, Farinella Z, Avola R, Beaudet A, Marchetti B. Basic fibroblast growth factor priming increases the responsiveness of immortalized hypothalamic luteinizing hormone releasing hormone neurones to neurotrophic factors. J Neuroendocrinol 2000; 12:941-59. [PMID: 11012835 DOI: 10.1046/j.1365-2826.2000.00554.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The participation of growth factors (GFs) in the regulation of luteinizing hormone releasing hormone (LHRH) neuronal function has recently been proposed, but little is known about the role played by GFs during early LHRH neurone differentiation. In the present study, we have used combined biochemical and morphological approaches to study the ability of a number of GFs normally expressed during brain development, including basic fibroblast growth factor (bFGF), epidermal growth factor (EGF), insulin and insulin-like growth factor I (IGF-I) to induce survival, differentiation, proliferation, and phenotypic expression of immortalized (GT1-1) LHRH neurones in vitro, at early (3-days in vitro, 3-DIV) and late (8-DIV) stages of neuronal differentiation. Comparison of GF-treated vs untreated neurones grown in serum-deprived (SD) medium demonstrated bFGF to be the most potent, and insulin the least active in promoting neuronal differentiation. Thus, at both 3-DIV and 8-DIV, but especially at 8-DIV, bFGF induced the greatest increase in the total length and number of LHRH processes/cell and in growth cone surface area. bFGF was also the most active at 3-DIV, and IGF-I at 8-DIV, in counteracting SD-induced cell death, whereas EGF was the most potent in increasing [3H]thymidine incorporation. All GFs studied decreased the spontaneous release of LHRH from GT1-1 cells when applied at 3-DIV or 8-DIV, except for insulin which was inactive at both time-points and bFGF which was inactive at 8-DIV. Pre-treatment of GT1-1 cells with a suboptimal ('priming') dose of bFGF for 12 h followed by application of the different GFs induced a sharp potentiation of the neurotrophic and proliferative effects of the latter and particularly of those of IGF-I. Moreover, bFGF priming counteracted EGF-induced decrease in LHRH release and significantly stimulated LHRH secretion following IGF-I or insulin application, suggesting that bFGF may sensitize LHRH neurones to differentiating effects of specific GFs during development.
Collapse
Affiliation(s)
- F Gallo
- Departments of Pharmacology, Medical School, University of Sassari, Pharmacology and Biochemistry, Medical School, University of Catania, Catania, Italy
| | | | | | | | | | | | | | | |
Collapse
|