1
|
Macauley SL, Horsch AD, Oterdoom M, Zheng MH, Stewart GR. The Effects of Transforming Growth Factor-β2 on Dopaminergic Graft Survival. Cell Transplant 2017; 13:245-52. [PMID: 15191162 DOI: 10.3727/000000004783984043] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Dopaminergic cell transplantation is a promising therapeutic approach for the treatment of Parkinson's disease, the potential of which is limited due to poor survival and low dopamine content within engrafted tissue. In this study, the ability of transforming growth factor-β2 (TGF-β2) to influence transplant survival was evaluated. Cell suspensions containing fetal rat ventral mesencephalon (VM) cells were incubated prior to surgery with vehicle (DPBS), varying concentrations of TGF-β2 (5–1000 ng/ml), or a pan-specific antibody against TGF-β (1D11, 100 ng/ml). VM cell suspensions (200,000 cells) were unilaterally implanted into the striatum of adult Sprague-Dawley rats (n = 5–11 animals/group). Following a 3-week survival period, small but viable VM grafts containing tyrosine hydroxylase-positive (TH+) neurons and fibers were present in all animals. Addition of TGF-β2 resulted in a steep, bell-shaped dose-response curve with a significant effect on TH+/dopamine cell survival. At 50 ng/ml TGF-β2, the number of surviving dopamine neurons was increased twofold compared with controls. Addition of TGF-β2 or 1D11 did not significantly influence graft volume. Further studies, possibly in combination with other neurotrophic factors, need to be performed to obtain a greater understanding of the effects of TGF-β on dopamine neurons and fetal VM cell engraftment.
Collapse
|
2
|
Neural Progenitor Cells Derived from Human Embryonic Stem Cells as an Origin of Dopaminergic Neurons. Stem Cells Int 2015; 2015:647437. [PMID: 26064138 PMCID: PMC4430666 DOI: 10.1155/2015/647437] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 04/11/2015] [Accepted: 04/14/2015] [Indexed: 12/14/2022] Open
Abstract
Human embryonic stem cells (hESCs) are able to proliferate in vitro indefinitely without losing their ability to differentiate into multiple cell types upon exposure to appropriate signals. Particularly, the ability of hESCs to differentiate into neuronal subtypes is fundamental to develop cell-based therapies for several neurodegenerative disorders, such as Alzheimer's disease, Huntington's disease, and Parkinson's disease. In this study, we differentiated hESCs to dopaminergic neurons via an intermediate stage, neural progenitor cells (NPCs). hESCs were induced to neural progenitor cells by Dorsomorphin, a small molecule that inhibits BMP signalling. The resulting neural progenitor cells exhibited neural bipolarity with high expression of neural progenitor genes and possessed multipotential differentiation ability. CBF1 and bFGF responsiveness of these hES-NP cells suggested their similarity to embryonic neural progenitor cells. A substantial number of dopaminergic neurons were derived from hES-NP cells upon supplementation of FGF8 and SHH, key dopaminergic neuron inducers. Importantly, multiple markers of midbrain neurons were detected, including NURR1, PITX3, and EN1, suggesting that hESC-derived dopaminergic neurons attained the midbrain identity. Altogether, this work underscored the generation of neural progenitor cells that retain the properties of embryonic neural progenitor cells. These cells will serve as an unlimited source for the derivation of dopaminergic neurons, which might be applicable for treating patients with Parkinson's disease.
Collapse
|
3
|
Wang T, Yuan W, Liu Y, Zhang Y, Wang Z, Zhou X, Ning G, Zhang L, Yao L, Feng S, Kong X. The role of the JAK-STAT pathway in neural stem cells, neural progenitor cells and reactive astrocytes after spinal cord injury. Biomed Rep 2014; 3:141-146. [PMID: 25798237 DOI: 10.3892/br.2014.401] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 10/16/2014] [Indexed: 12/18/2022] Open
Abstract
Patients with spinal cord injuries can develop severe neurological damage and dysfunction, which is not only induced by primary but also by secondary injuries. As an evolutionarily conserved pathway of eukaryotes, the JAK-STAT pathway is associated with cell growth, survival, development and differentiation; activation of the JAK-STAT pathway has been previously reported in central nervous system injury. The JAK-STAT pathway is directly associated with neurogenesis and glia scar formation in the injury region. Following injury of the axon, the overexpression and activation of STAT3 is exhibited specifically in protecting neurons. To investigate the role of the JAK-STAT pathway in neuroprotection, we summarized the effect of JAK-STAT pathway in the following three sections: Firstly, the modulation of JAK-STAT pathway in proliferation and differentiation of neural stem cells and neural progenitor cells is discussed; secondly, the time-dependent effect of JAK-STAT pathway in reactive astrocytes to reveal their capability of neuroprotection is revealed and lastly, we focus on how the astrocyte-secretory polypeptides (astrocyte-derived cytokines and trophic factors) accomplish neuroprotection via the JAK-STAT pathway.
Collapse
Affiliation(s)
- Tianyi Wang
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China ; Department of Orthopedics, The 266th Hospital of the Chinese People's Liberation Army, Chengde, Hebei 067000, P.R. China
| | - Wenqi Yuan
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Yong Liu
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Yanjun Zhang
- Department of Orthopedics, Capital Medical University Luhe Hospital, Beijing 100000, P.R. China
| | - Zhijie Wang
- Department of Paediatric Internal Medicine, Affiliated Hospital of Chengde Medical College, Chengde, Hebei 067000, P.R. China
| | - Xianhu Zhou
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Guangzhi Ning
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Liang Zhang
- Department of Orthopedics, The Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Liwei Yao
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Shiqing Feng
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Xiaohong Kong
- School of Medicine, Nankai University, Tianjin 300071, P.R. China
| |
Collapse
|
4
|
Establishment of a survival and toxic cellular model for Parkinson's disease from chicken mesencephalon. Neurotox Res 2012; 24:119-29. [PMID: 23238634 PMCID: PMC3691473 DOI: 10.1007/s12640-012-9367-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Revised: 11/28/2012] [Accepted: 11/29/2012] [Indexed: 12/21/2022]
Abstract
Cellular models for Parkinson’s disease (PD) represent a fast and efficient tool in the screening for drug candidates and factors involved in the disease pathogenesis. The objective of this study was to establish and characterize a survival and toxic cellular model for PD by culturing dopaminergic neurons from embryonic chicken ventral midbrain. We show that as in rodents, the common neurotrophic factors—brain-derived neurotrophic factor (BDNF), glial cell line-derived neurotrophic factor (GDNF), and fibroblast growth factor 2 (FGF2)—are able to support the survival of chicken midbrain dopaminergic neurons. Furthermore, after treatment with MPP+ or rotenone as in vitro models for PD, the number of tyrosine hydroxylase-positive cells decreased drastically. This effect could be significantly rescued by treatment with BDNF or GDNF. Together, our results indicate that mechanisms of neuroprotection of dopaminergic neurons are conserved between chicken and mammals. This supports the use of primary culture from chicken embryonic midbrain as a suitable tool for the study of neuroprotection in vitro.
Collapse
|
5
|
Sleeman IJ, Boshoff EL, Duty S. Fibroblast growth factor-20 protects against dopamine neuron loss in vitro and provides functional protection in the 6-hydroxydopamine-lesioned rat model of Parkinson's disease. Neuropharmacology 2012; 63:1268-77. [PMID: 22971544 DOI: 10.1016/j.neuropharm.2012.07.029] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Revised: 06/25/2012] [Accepted: 07/15/2012] [Indexed: 11/25/2022]
Abstract
Fibroblast growth factor-20 (FGF-20) has been shown to protect dopaminergic neurons against a range of toxic insults in vitro, through activation of fibroblast growth factor receptor 1 (FGFR1). This study set out to examine whether FGF-20 also displayed protective efficacy in the unilateral, 6-hydroxydopamine (6-OHDA) lesion rat model of Parkinson's disease. Initial studies demonstrated that, in embryonic ventral mesencephalic (VM) cultures, FGFR1 was expressed on tyrosine hydroxylase (TH)-positive neurons and that, in line with previous data, FGF-20 (100 and 500 ng/ml) almost completely protected these TH-positive neurons against 6-OHDA-induced toxicity. Co-localisation of FGFR1 and TH staining was also demonstrated in the substantia nigra pars compacta (SNpc) of naïve adult rat brain. In animals subject to 6-OHDA lesion of the nigrostriatal tract, supra-nigral infusion of FGF-20 (2.5 μg/day) for 6 days post-lesion gave significant protection (∼40%) against the loss of TH-positive cells in the SNpc and the loss of striatal TH immunoreactivity. This protection of the nigrostriatal tract was accompanied by a significant preservation of gross locomotion and fine motor movements and reversal of apomorphine-induced contraversive rotations, although forelimb akinesia, assessed using cylinder test reaching, was not improved. These results support a role for FGF-20 in preserving dopamine neuron integrity and some aspects of motor function in a rodent model of Parkinson's disease (PD) and imply a potential neuroprotective role for FGF-20 in this disease.
Collapse
Affiliation(s)
- Isobel J Sleeman
- King's College London, Wolfson Centre for Age-Related Diseases, Guy's Campus, London SE1 1UL, UK
| | | | | |
Collapse
|
6
|
Shi B, Deng L, Shi X, Dai S, Zhang H, Wang Y, Bi J, Guo M. The enhancement of neural stem cell survival and growth by coculturing with expanded Sertoli cells in vitro. Biotechnol Prog 2011; 28:196-205. [PMID: 22109810 DOI: 10.1002/btpr.720] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Revised: 08/20/2011] [Indexed: 12/19/2022]
Abstract
Sertoli cells (SCs) have been described as the "nurse cells" of testis to provide essential growth factors and to create a proper environment for the development of other cells (e.g., germinal and neural stem cell). However, the physiological functions of the SCs obtained from different culture conditions are different in a coculturing system, and thus the optimal SC culturing condition should be investigated in vitro. In this paper, primary Sertoli cells were isolated from a 12-day-old mouse and expanded in two different culture conditions: a two dimensional (2D) plastic tissue disc and a three dimensional (3D) microcarrier culture system. They were then cocultured with neural stem cells (NSCs) isolated from 14-day-old mouse embryos. The metabolic activities of SCs(2D) (SCs in 2D) and SCs(3D) (SCs in 3D) and the amount of proteins secreted from two culturing systems were compared. The results show that the metabolic activity and the amount of secreted proteins from SCs(3D) were higher than both from SCs(2D). Three coculturing groups: NSCs+SC(2D), NSCs+SC(3D), and NSCs +SC-conditioned medium (SCCM, control group) were also compared regarding cell morphology and the numbers of neurons, neural outgrowths and neurospheres. The quantity of neurons, neural outgrowths and neurospheres were the highest in the NSCs+SC(3D) group. SCs cultured in the 3D system had a strong trophic effect on NSCs and enhanced their survival and growth. Besides, the mRNA of trophic and nutritive factors such as Glial-cell-line-derived neurotrophic factor (GDNF) and Interleukin-1 α (IL-1 α) secreted by the SCs from both 2D and 3D culture system were analyzed by real time-PCR and gel assay. The mRNA transcription of GDNF and IL-1α is more apparent in the 3D culture system than that from the 2D one. The coculturing system of NSCs+SC(3D) is a promising candidate for future neural stem cell transplantation.
Collapse
Affiliation(s)
- Bingyang Shi
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China
| | | | | | | | | | | | | | | |
Collapse
|
7
|
Werner S, Unsicker K, von Bohlen und Halbach O. Fibroblast growth factor-2 deficiency causes defects in adult hippocampal neurogenesis, which are not rescued by exogenous fibroblast growth factor-2. J Neurosci Res 2011; 89:1605-17. [PMID: 21800348 DOI: 10.1002/jnr.22680] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2010] [Revised: 03/29/2011] [Accepted: 03/31/2011] [Indexed: 12/17/2022]
Abstract
Neurogenesis within the adult brain is restricted to selected areas, one of which is the dentate gyrus (DG). Several growth factors have been reported to affect neurogenesis in the adult DG. However, a role of fibroblast growth factor-2 (FGF-2) in adult hippocampal neurogenesis has not been firmly established. We have analyzed neurogenesis in the DG using in vivo and in vitro approaches. FGF-2(-/-) mice revealed no alterations in the number of proliferating cells but a significant decrease in the numbers of newly generated neurons. Moreover, FGF-2 added to hippocampal slice cultures from FGF-2(-/-) mice was unable to rescue the phenotype. Although an increase in death of neurogenic cells in the FGF-2-deficient DG could not be specifically demonstrated, there was a massive increase in global cell death in FGF-2(-/-) hippocampal slice cultures compared with slices from wild-type mice. Cell death could not be prevented by addition of FGF-2. Neutralization of endogenous FGF-2 in hippocampal slices did not interfere with neurogenesis in a short-term paradigm. Together, our data suggest that FGF-2 is essentially required for maturation of new neurons in adult hippocampal neurogenesis but is likely to operate synergistically in combination with other mechanisms/growth factors.
Collapse
Affiliation(s)
- Sandra Werner
- Interdisciplinary Center for Neurosciences, Department of Neuroanatomy, Heidelberg, Germany.
| | | | | |
Collapse
|
8
|
Harness JV, Turovets NA, Seiler MJ, Nistor G, Altun G, Agapova LS, Ferguson D, Laurent LC, Loring JF, Keirstead HS. Equivalence of conventionally-derived and parthenote-derived human embryonic stem cells. PLoS One 2011; 6:e14499. [PMID: 21249129 PMCID: PMC3017547 DOI: 10.1371/journal.pone.0014499] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2010] [Accepted: 11/17/2010] [Indexed: 12/31/2022] Open
Abstract
Background As human embryonic stem cell (hESC) lines can be derived via multiple means, it is important to determine particular characteristics of individual lines that may dictate the applications to which they are best suited. The objective of this work was to determine points of equivalence and differences between conventionally-derived hESC and parthenote-derived hESC lines (phESC) in the undifferentiated state and during neural differentiation. Methodology/Principal Findings hESC and phESC were exposed to the same expansion conditions and subsequent neural and retinal pigmented epithelium (RPE) differentiation protocols. Growth rates and gross morphology were recorded during expansion. RTPCR for developmentally relevant genes and global DNA methylation profiling were used to compare gene expression and epigenetic characteristics. Parthenote lines proliferated more slowly than conventional hESC lines and yielded lower quantities of less mature differentiated cells in a neural progenitor cell (NPC) differentiation protocol. However, the cell lines performed similarly in a RPE differentiation protocol. The DNA methylation analysis showed similar general profiles, but the two cell types differed in methylation of imprinted genes. There were no major differences in gene expression between the lines before differentiation, but when differentiated into NPCs, the two cell types differed in expression of extracellular matrix (ECM) genes. Conclusions/Significance These data show that hESC and phESC are similar in the undifferentiated state, and both cell types are capable of differentiation along neural lineages. The differences between the cell types, in proliferation and extent of differentiation, may be linked, in part, to the observed differences in ECM synthesis and methylation of imprinted genes.
Collapse
Affiliation(s)
- Julie V. Harness
- Reeve-Irvine Research Center, Sue and Bill Gross Stem Cell Research Center, Department of Anatomy and Neurobiology, School of Medicine, University of California at Irvine, Irvine, California, United States of America
| | - Nikolay A. Turovets
- International Stem Cell Corporation, Oceanside, California, United States of America
| | - Magdalene J. Seiler
- Reeve-Irvine Research Center, Sue and Bill Gross Stem Cell Research Center, Department of Anatomy and Neurobiology, School of Medicine, University of California at Irvine, Irvine, California, United States of America
| | - Gabriel Nistor
- Reeve-Irvine Research Center, Sue and Bill Gross Stem Cell Research Center, Department of Anatomy and Neurobiology, School of Medicine, University of California at Irvine, Irvine, California, United States of America
| | - Gulsah Altun
- Center for Regenerative Medicine, Scripps Research Institute, La Jolla, California, United States of America
| | - Larissa S. Agapova
- International Stem Cell Corporation, Oceanside, California, United States of America
| | - David Ferguson
- Reeve-Irvine Research Center, Sue and Bill Gross Stem Cell Research Center, Department of Anatomy and Neurobiology, School of Medicine, University of California at Irvine, Irvine, California, United States of America
| | - Louise C. Laurent
- Center for Regenerative Medicine, Scripps Research Institute, La Jolla, California, United States of America
| | - Jeanne F. Loring
- Center for Regenerative Medicine, Scripps Research Institute, La Jolla, California, United States of America
| | - Hans S. Keirstead
- Reeve-Irvine Research Center, Sue and Bill Gross Stem Cell Research Center, Department of Anatomy and Neurobiology, School of Medicine, University of California at Irvine, Irvine, California, United States of America
- * E-mail:
| |
Collapse
|
9
|
Schindowski K, von Bohlen und Halbach O, Strelau J, Ridder DA, Herrmann O, Schober A, Schwaninger M, Unsicker K. Regulation of GDF-15, a distant TGF-β superfamily member, in a mouse model of cerebral ischemia. Cell Tissue Res 2010; 343:399-409. [PMID: 21128084 PMCID: PMC3032194 DOI: 10.1007/s00441-010-1090-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Accepted: 11/04/2010] [Indexed: 11/20/2022]
Abstract
GDF-15 is a novel distant member of the TGF-β superfamily and is widely distributed in the brain and peripheral nervous system. We have previously reported that GDF-15 is a potent neurotrophic factor for lesioned dopaminergic neurons in the substantia nigra, and that GDF-15-deficient mice show progressive postnatal losses of motor and sensory neurons. We have now investigated the regulation of GDF-15 mRNA and immunoreactivity in the murine hippocampal formation and selected cortical areas following an ischemic lesion by occlusion of the middle cerebral artery (MCAO). MCAO prominently upregulates GDF-15 mRNA in the hippocampus and parietal cortex at 3 h and 24 h after lesion. GDF-15 immunoreactivity, which is hardly detectable in the unlesioned brain, is drastically upregulated in neurons identified by double-staining with NeuN. NeuN staining reveals that most, if not all, neurons in the granular layer of the dentate gyrus and pyramidal layers of the cornu ammonis become GDF-15-immunoreactive. Moderate induction of GDF-15 immunoreactivity has been observed in a small number of microglial cells identified by labeling with tomato lectin, whereas astroglial cells remain GDF-15-negative after MCAO. Comparative analysis of the size of the infarcted area after MCAO in GDF-15 wild-type and knockout mice has failed to reveal significant differences. Together, our data substantiate the notion that GDF-15 is prominently upregulated in the lesioned brain and might be involved in orchestrating post-lesional responses other than the trophic support of neurons.
Collapse
Affiliation(s)
- Katharina Schindowski
- Institute for Pharmaceutical Biotechnology, University of Applied Science Biberach, Biberach/Riss, Germany
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Gonzalez-Aparicio R, Flores JA, Fernandez-Espejo E. Antiparkinsonian trophic action of glial cell line-derived neurotrophic factor and transforming growth factor β1 is enhanced after co-infusion in rats. Exp Neurol 2010; 226:136-47. [DOI: 10.1016/j.expneurol.2010.08.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2010] [Revised: 07/22/2010] [Accepted: 08/10/2010] [Indexed: 02/03/2023]
|
11
|
Hamby ME, Sofroniew MV. Reactive astrocytes as therapeutic targets for CNS disorders. Neurotherapeutics 2010; 7:494-506. [PMID: 20880511 PMCID: PMC2952540 DOI: 10.1016/j.nurt.2010.07.003] [Citation(s) in RCA: 259] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2010] [Revised: 07/19/2010] [Accepted: 07/20/2010] [Indexed: 12/30/2022] Open
Abstract
Reactive astrogliosis has long been recognized as a ubiquitous feature of CNS pathologies. Although its roles in CNS pathology are only beginning to be defined, genetic tools are enabling molecular dissection of the functions and mechanisms of reactive astrogliosis in vivo. It is now clear that reactive astrogliosis is not simply an all-or-nothing phenomenon but, rather, is a finely gradated continuum of molecular, cellular, and functional changes that range from subtle alterations in gene expression to scar formation. These changes can exert both beneficial and detrimental effects in a context-dependent manner determined by specific molecular signaling cascades. Dysfunction of either astrocytes or the process of reactive astrogliosis is emerging as an important potential source of mechanisms that might contribute to, or play primary roles in, a host of CNS disorders via loss of normal or gain of abnormal astrocyte activities. A rapidly growing understanding of the mechanisms underlying astrocyte signaling and reactive astrogliosis has the potential to open doors to identifying many molecules that might serve as novel therapeutic targets for a wide range of neurological disorders. This review considers general principles and examines selected examples regarding the potential of targeting specific molecular aspects of reactive astrogliosis for therapeutic manipulations, including regulation of glutamate, reactive oxygen species, and cytokines.
Collapse
Affiliation(s)
- Mary E. Hamby
- grid.19006.3e0000000096326718Department of Neurobiology, David Geffen School of Medicine, University of California, 90095 Los Angeles, California
| | - Michael V. Sofroniew
- grid.19006.3e0000000096326718Department of Neurobiology, David Geffen School of Medicine, University of California, 90095 Los Angeles, California
| |
Collapse
|
12
|
Asano Y, Koishi K, Frugier T, McLennan IS. Mice with disrupted TGFbeta signaling have normal cerebella development, but exhibit facial dysmorphogenesis and strain-dependent deficits in their body wall. Cell Mol Neurobiol 2009; 29:621-33. [PMID: 19214740 PMCID: PMC11505784 DOI: 10.1007/s10571-009-9354-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2008] [Accepted: 01/22/2009] [Indexed: 01/07/2023]
Abstract
The transforming growth factor betas (TGFbetas) are context-dependent regulators of neurons in vitro, but their physiological functions in the brain are unclear. Haploinsufficiency of either Tgfbeta1 or Tgfbeta2 leads to age-related deterioration of neurons, but the development of the brain is normal in the full absence of either of these genes. However, some individuals with mis-sense mutations of TGFbeta receptors are mentally retarded, suggesting that the TGFbeta isoforms can compensate for each other during brain development. This possibility was tested by generating mice (NSE x PTR) with neuron-specific expression of a dominant-negative inhibitor of TGFbeta signaling. The NSE x PTR mice with a FVBxC57Bl/6 genetic background were viable and developed normally despite strong neuronal expression of the inhibitor of TGFbeta signaling. Their cerebella were of normal size and contained normal numbers of neurons. When the genetic background of the mice was changed to C57BL/6, the phenotype of the mice became neonatal lethal, with the neonates exhibiting various malformations. The malformations correlated with sites of non-neuronal expression of the transgenes and included facial dysmorphogenesis, incomplete closure of the ventral body wall and absence of intestinal motility. The C57BL/6 Tgfbm1-3 alleles, which modulate the phenotype of Tgfbeta1(-/-) mice, were not major determinants of the NSE x PTR phenotype. The data suggest that the development of the cerebellum is insensitive to the level of TGFbeta signaling, although this may be dependent on the genetic background.
Collapse
Affiliation(s)
- Yoshiya Asano
- Department of Anatomy and Structural Biology, Neuromuscular Research Group, Otago School of Medical Sciences, University of Otago, P.O. Box 913, Dunedin, New Zealand
- Present Address: Department of Neuroanatomy, Cell Biology and Histology, Hirosaki University, Graduate School of Medicine, Hirosaki, 036-8562 Japan
| | - Kyoko Koishi
- Department of Anatomy and Structural Biology, Neuromuscular Research Group, Otago School of Medical Sciences, University of Otago, P.O. Box 913, Dunedin, New Zealand
| | - Tony Frugier
- Department of Anatomy and Structural Biology, Neuromuscular Research Group, Otago School of Medical Sciences, University of Otago, P.O. Box 913, Dunedin, New Zealand
- Present Address: National Trauma Research Institute, The Alfred Hospital, 89 Commercial Road, Melbourne, VIC 3004 Australia
| | - Ian S. McLennan
- Department of Anatomy and Structural Biology, Neuromuscular Research Group, Otago School of Medical Sciences, University of Otago, P.O. Box 913, Dunedin, New Zealand
| |
Collapse
|
13
|
Morphophysiology of the Zuckerkandl's paraganglion: effects of dexamethasone and aging. Neurobiol Aging 2009; 31:2115-27. [PMID: 19167134 DOI: 10.1016/j.neurobiolaging.2008.12.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2008] [Revised: 11/26/2008] [Accepted: 12/02/2008] [Indexed: 11/23/2022]
Abstract
The extra-adrenal Zuckerkandl's paraganglion is used as a source of chromaffin cells for transplantation in parkinsonian animals. Aging can affect its viability, and this tissue needs further characterization for improving grafting procedures. The objectives were: (i) to compare the main morpho-functional characteristics of prepubertal and old Zuckerkandl's paraganglion (ZP), and (ii) to discern phenotypic changes after sub-chronic dexamethasone treatment in extra-adrenal tissue of prepubertal rats. For these purposes, immunostaining methods, stereology, voltammetry, cell culture, Western blotting, and ELISA were employed. The findings revealed that all paraganglia were composed of mesenchymal tissue and chromaffin cells. In prepubertal rats, chromaffin cells are arranged as large or small clusters. Large clusters (also known as "cell nests") contain densely packed chromaffin cells, and they are seen as fascicles in longitudinal sections. In old paraganglia, cell nests disappear, and chromaffin cells are found to be arranged as small cell clusters or dispersed throughout the mesenchyma. Paraganglionic chromaffin cells possess a rounded morphology with diameter ranging from 12 to 15 μm, with intracytoplasmic granules (100-500 nm in diameter) containing catecholamines. Prepubertal and old ZP chromaffin cells are mostly noradrenergics, and a few of them are dopaminergics. Aging reduces the amount of chromaffin tissue (28% in adult rats vs. 11% in old animals, both in relation to total volume of the paraganglion), and induces the presence of adrenergic cells and adrenaline. Both prepubertal and old cells express the neurotrophic factors GDNF and TGF-β₁, aging leading to reduced levels of both growth factors. Dexamethasone (50 μg/kg daily, 5 days) leads to the expression of phenylethanolamine-N-methyl-transferase in prepubertal paraganglia, and to a higher content and release of adrenaline.
Collapse
|
14
|
Molecular and cellular determinants for generating ES-cell derived dopamine neurons for cell therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2009; 651:112-23. [PMID: 19731556 DOI: 10.1007/978-1-4419-0322-8_11] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Embryonic stem (ES) cells can generate midbrain dopaminergic (DA) neuronal phenotypes in vitro and have been successfully applied to restore function in animal models of Parkinson's disease (PD). How can we best integrate our growinginsight into the regulatory cascade of transcription factors guiding midbrain specification to further improve the in vitro differentiation of midbrain DA neurons for cell therapy of PD? To characterize the differentiation of authentic DA neurons in vitro, expression patterns of the numerous midbrain-characteristic markers need to be investigated. When using forced gene expression, such factors have to be closely monitored to avoid generation of nonphysiological cell types. Fluorescent markers such as Pitx3-GFP, TH-GFP, Sox1-GFP or surface antigens have proven useful for elimination of unwanted cell types by cell sorting, thereby averting tumors and increasing the DA fraction for transplantation studies. The importance of appropriate timing during application of extrinsic factors and the influence of cell-cell interactions in the dish has to be taken into account. This conceptual synopsis outlines current objectives, progress, but also challenges, in deriving midbrain DA neurons from pluripotent stem cells for clinical and scientific applications.
Collapse
|
15
|
Saito M, Terada M, Kawata T, Ito H, Shigematsu N, Kromkhun P, Yokosuka M, Saito TR. Effects of single or repeated administrations of methamphetamine on immune response in mice. Exp Anim 2008; 57:35-43. [PMID: 18256517 DOI: 10.1538/expanim.57.35] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
The present study aimed to clarify the connection between immune responses and the administration frequency of methamphetamine (MAP) in male and female mice. Male and female ddY mice were given single or multiple (repeated for 10 days) intraperitoneal injections of MAP (5.0 mg/kg/day). The following immune parameters were examined; the number of leukocytes in peripheral blood and the proliferative activity (phytohemagglutinin;PHA, lipopolysaccharide; LPS response) and natural killer (NK) cell activity in splenic lymphocytes. Further, the differences in metabolic function in the spleen in response to MAP (and its metabolite amphetamine) in male and female mice were measured by gas chromatography. The results of the present study were that; 1) single and repeated MAP injections reduced leukocytes; 2) single MAP injection increased the proliferative response of splenic lymphocytes to PHA stimulation in only male mice, but the response to LPS stimulation was slightly increased in both male and female mice; 3) single and repeated MAP injections reduced NK cell activity of splenic lymphocytes, and especially in female mice with 5 injections of MAP; 4) with 10 MAP injections the NK cell activity and leukocytes recovered to the level of controls; and 5) the metabolic activity of MAP was reduced in female mice treated acutely with MAP in comparison to male mice. These results appear to indicate that immune responses to MAP were involved in the different results shown for administration frequency, sex difference and metabolic process of MAP.
Collapse
Affiliation(s)
- Masayoshi Saito
- Department of Radiology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Galan-Rodriguez B, del-Marco A, Flores J, Ramiro-Fuentes S, Gonzalez-Aparicio R, Tunez I, Tasset I, Fernandez-Espejo E. Grafts of extra-adrenal chromaffin cells as aggregates show better survival rate and regenerative effects on parkinsonian rats than dispersed cell grafts. Neurobiol Dis 2008; 29:529-42. [DOI: 10.1016/j.nbd.2007.11.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2007] [Revised: 10/29/2007] [Accepted: 11/17/2007] [Indexed: 11/28/2022] Open
|
17
|
Aigner L, Bogdahn U. TGF-beta in neural stem cells and in tumors of the central nervous system. Cell Tissue Res 2007; 331:225-41. [PMID: 17710437 DOI: 10.1007/s00441-007-0466-7] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2007] [Accepted: 07/04/2007] [Indexed: 10/22/2022]
Abstract
Mechanisms that regulate neural stem cell activity in the adult brain are tightly coordinated. They provide new neurons and glia in regions associated with high cellular and functional plasticity, after injury, or during neurodegeneration. Because of the proliferative and plastic potential of neural stem cells, they are currently thought to escape their physiological control mechanisms and transform to cancer stem cells. Signals provided by proteins of the transforming growth factor (TGF)-beta family might represent a system by which neural stem cells are controlled under physiological conditions but released from this control after transformation to cancer stem cells. TGF-beta is a multifunctional cytokine involved in various physiological and patho-physiological processes of the brain. It is induced in the adult brain after injury or hypoxia and during neurodegeneration when it modulates and dampens inflammatory responses. After injury, although TGF-beta is neuroprotective, it may limit the self-repair of the brain by inhibiting neural stem cell proliferation. Similar to its effect on neural stem cells, TGF-beta reveals anti-proliferative control on most cell types; however, paradoxically, many brain tumors escape from TGF-beta control. Moreover, brain tumors develop mechanisms that change the anti-proliferative influence of TGF-beta into oncogenic cues, mainly by orchestrating a multitude of TGF-beta-mediated effects upon matrix, migration and invasion, angiogenesis, and, most importantly, immune escape mechanisms. Thus, TGF-beta is involved in tumor progression. This review focuses on TGF-beta and its role in the regulation and control of neural and of brain-cancer stem cells.
Collapse
Affiliation(s)
- Ludwig Aigner
- Department of Neurology, University of Regensburg, Universitätsstrasse 84, 93053, Regensburg, Germany.
| | | |
Collapse
|
18
|
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
|
19
|
Adriani W, Leo D, Guarino M, Natoli A, Di Consiglio E, De Angelis G, Traina E, Testai E, Perrone-Capano C, Laviola G. Short-Term Effects of Adolescent Methylphenidate Exposure on Brain Striatal Gene Expression and Sexual/Endocrine Parameters in Male Rats. Ann N Y Acad Sci 2006; 1074:52-73. [PMID: 17105903 DOI: 10.1196/annals.1369.005] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Exposure to methylphenidate (MPH) during adolescence is the elective therapy for attention deficit/hyperactivity disorder (ADHD) children, but raises major concerns for public health, due to possibly persistent neurobehavioral changes. Rats (30- to 44-days old) were administered MPH (2 mg/kg, i.p once daily) or saline (SAL). At the end of the treatment we collected plasma, testicular, liver, and brain (striatum) samples. The testes and liver were used to evaluate conventional reproductive and metabolic endpoints. Testes of MPH-exposed rats weighed more and contained an increased quantity of sperm, whereas testicular levels of testosterone (TST) were markedly decreased. The MPH treatment exerted an inductive effect on enzymatic activity of TST hydroxylases, resulting in increased hepatic TST catabolism. These findings suggest that subchronic MPH exposure in adolescent rats could have a trophic action on testis growth and a negative impact on TST metabolism. We have analyzed striatal gene expression profiles as a consequence of MPH exposure during adolescence, using microarray technology. More than 700 genes were upregulated in the striatum of MPH-treated rats (foldchange >1.5). A first group of genes were apparently involved in migration of immature neural/glial cells and/or growth of novel axons. These genes include matrix proteases (ADAM-1, MMP14), their inhibitors (TIMP-2, TIMP-3), the hyaluronan-mediated motility receptor (RHAMM), and growth factors (transforming growth factor-beta3 [TGF-beta3] and fibroblast growth factor 14 [FGF14]). A second group of genes were suggestive of active axonal myelination. These genes mediate survival of immature cells after contact with newly produced axonal matrix (laminin B1, collagens, integrin alpha 6) and stabilization of myelinating glia-axon contacts (RAB13, contactins 3 and 4). A third group indicated the appearance and/or upregulation of mature processes. The latter included genes for: K+ channels (TASK-1, TASK-5), intercellular junctions (connexin30), neurotransmitter receptors (adrenergic alpha 1B, kainate 2, serotonin 7, GABA-A), as well as major proteins responsible for their transport and/or anchoring (Homer 1, MAGUK MPP3, Shank2). All these genes were possibly involved in synaptic plasticity, namely the formation, maturation, and stabilization of new neural connections within the striatum. MPH treatment seems to potentiate synaptic plasticity, which is an age-dependent developmental phenomenon that adolescent rats are very likely to show, compared to adults. Our observations suggest that adolescent MPH exposure causes only transient changes in reproductive and hormonal parameters, and a more enduring enhancement of neurobehavioral plasticity.
Collapse
Affiliation(s)
- Walter Adriani
- Department of Cell Biology & Neurosciences, Behavioural Neuroscience Section, Istituto Superiore di Sanità, viale Regina Elena 299, I-00161 Roma, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Shamekh R, Mallery J, Newcomb J, Hushen J, Saporta S, Cameron DF, Sanberg CD, Sanberg PR, Willing AE. Enhancing tyrosine hydroxylase expression and survival of fetal ventral mesencephalon neurons with rat or porcine Sertoli cells in vitro. Brain Res 2006; 1096:1-10. [PMID: 16780819 DOI: 10.1016/j.brainres.2006.04.058] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2005] [Revised: 03/29/2006] [Accepted: 04/03/2006] [Indexed: 10/24/2022]
Abstract
Sertoli cells (SCs) are testis-derived cells that secrete trophic factors important for the development of germ cells. Both porcine and rat SCs have been used as graft facilitators - neonatal porcine SCs to support islets in diabetes and 15-day-old rat SCs to enhance dopaminergic neuron transplants in Parkinson's disease models. However, there has never been a study examining the optimal SCs preparation to enhance tyrosine hydroxylase expression in the ventral mesencephalon (VM) neuron. The aim of this study was to compare the ability of both rat and porcine SCs to enhance tyrosine hydroxylase expression (TH) and neuronal survival at the same postnatal developmental ages. The SCs were isolated from 1-, 9-, or 15-day-old rat, or neonate (2-5 days), 2-month, or 4-month-old pig, and co-cultured with VM tissue from 13.5-day-old embryos. Our results showed that VM neurons co-cultured with SCs dispersed over the culture plate and had extensive neuritic outgrowth, while VM neurons cultured alone tended to cluster together forming a mass of cells with limited neurite outgrowth. TH expression was significantly increased when VM neurons were co-cultured with 15-day rat SCs or 2-month pig SCs but not when the cells were co-cultured with other ages of SCs. This suggests that secretion of trophic factors by SCs varies according to the developmental age, and it is critical for the success of graft facilitation that SCs from the appropriate age and species be used.
Collapse
Affiliation(s)
- Rania Shamekh
- Department of Neurosurgery, Center of Excellence for Aging and Brain Repair, University of South Florida, Tampa, 33612, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Fumagalli F, Pasquale L, Racagni G, Riva MA. Dynamic regulation of fibroblast growth factor 2 (FGF-2) gene expression in the rat brain following single and repeated cocaine administration. J Neurochem 2006; 96:996-1004. [PMID: 16412094 DOI: 10.1111/j.1471-4159.2005.03627.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Administration of drugs of abuse can produce long-lasting effects on brain function, which involve modifications at neurotransmitter level as well as changes in proteins important for structural alterations of selected brain regions. The contribution of trophic factors in these events has so far been underestimated. Here, we demonstrate that a single cocaine injection selectively up-regulated fibroblast growth factor 2 (FGF-2) mRNA levels in the striatum and prefrontal cortex within 2 h, an effect that vanished by 24 h. However, prolonged exposure (5 or 14 days) to cocaine treatment produced an enduring elevation of FGF-2 mRNA levels that was evident 72 h after the last injection in the prefrontal cortex and could even persist for 14 days in the striatum, raising the possibility that cocaine treatment primes the brain, resulting in longer-lasting FGF-2 up-regulation in regions that are highly innervated by dopaminergic projections. The expression of FGF-2 was also significantly increased in the midbrain following acute or 5-day injection, suggesting that modulation of FGF-2 biosynthesis in dopamine-producing cells occurs only during early stages of cocaine exposure. Our results point to important mechanistic conclusions as to how cocaine alters FGF-2 expression. Whereas cocaine-induced changes in FGF-2 gene expression following a single injection could be ascribed to increased release of transmitters (mainly dopamine), enhanced FGF-2 gene expression following repeated administration identifies the trophic factor as part of the adaptive changes set in motion by cocaine.
Collapse
Affiliation(s)
- Fabio Fumagalli
- Center of Neuropharmacology, Department of Pharmacological Sciences, University of Milan, Milan, Italy
| | | | | | | |
Collapse
|
22
|
Fumagalli F, Bedogni F, Slotkin TA, Racagni G, Riva MA. Prenatal stress elicits regionally selective changes in basal FGF-2 gene expression in adulthood and alters the adult response to acute or chronic stress. Neurobiol Dis 2005; 20:731-7. [PMID: 15967670 DOI: 10.1016/j.nbd.2005.05.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2005] [Revised: 03/16/2005] [Accepted: 05/02/2005] [Indexed: 10/25/2022] Open
Abstract
Exposure to stress during pregnancy influences the trajectory of brain development resulting in permanent alterations that may contribute to increased susceptibility to subsequent cognitive or neuropsychiatric disorders. In this manuscript, we examined the effects of prenatal stress on the expression of basic fibroblast growth factor (FGF-2), an important molecular regulator of development and plasticity, in adult male rats under basal conditions as well as in response to acute or chronic stress. Baseline FGF-2 mRNA levels were differentially influenced by gestational stress in a variety of brain regions, with significant decreases in prefrontal cortex and increases in entorhinal cortex and striatum. By itself, postnatal stress similarly decreased trophic factor expression in prefrontal cortex while evoking stimulation elsewhere. Gestational stress altered the pattern of FGF-2 expression in response to adult stress, completely reversing the pattern in the prefrontal cortex (stimulatory instead of inhibitory), blunting the response in the entorhinal cortex and desensitizing the response in the striatum. These effects point to a unique interference of chronic prenatal stress with both ongoing FGF-2 expression and its responses to subsequent stressors, lasting into adulthood. Given the multifaceted role of FGF-2 in synaptic development, maintenance and plasticity, these data provide detailed mechanistic evidence as to how prenatal stress elicits lifelong effects on synaptic function. The abnormal modulation of FGF-2 gene expression in specific brain regions in response to subsequent stress in adulthood may impair the normal adaptive responses of the cell to challenging situations.
Collapse
Affiliation(s)
- Fabio Fumagalli
- Center of Neuropharmacology, Department of Pharmacological Sciences and Center of Excellence on Neurodegenerative Diseases (CEND), University of Milan, Via Balzaretti 9, 20133 Milan, Italy
| | | | | | | | | |
Collapse
|
23
|
Gomes FCA, Sousa VDO, Romão L. Emerging roles for TGF-beta1 in nervous system development. Int J Dev Neurosci 2005; 23:413-24. [PMID: 15936920 DOI: 10.1016/j.ijdevneu.2005.04.001] [Citation(s) in RCA: 123] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2004] [Revised: 04/14/2005] [Accepted: 04/14/2005] [Indexed: 01/07/2023] Open
Abstract
Transforming growth factor betas (TGF-betas) are known as multifunctional growth factors, which participate in the regulation of key events of development, disease and tissue repair. In central nervous system (CNS), TGF-beta1 has been widely recognized as an injury-related cytokine, specially associated with astrocyte scar formation in response to brain injury. TGF-betas family is represented by three isoforms: TGF-beta1, -beta 2 and -beta 3, all produced by both glial and neuronal cells. They are involved in essential tissue functions, including cell-cycle control, regulation of early development and differentiation, neuron survival and astrocyte differentiation. TGF-beta signaling is mediated mainly by two serine threonine kinase receptors, TGFRI and TGFRII, which activate Smad 2/3 and Smad 4 transcription factors. Phosphorylation and activation of these proteins is followed by formation of Smad 2/3-4 complex, which translocates to the nucleus regulating transcriptional responses to TGF-beta. Very few data are available concerning the intracellular pathway required for the effect of TGF-beta in brain cells. Recently, emerging data on TGF-beta1 and its signaling molecules have been suggesting that besides its role in brain injury, TGF-beta1 might be a crucial regulator of CNS development. In this review, we will focus on TGF-betas members, specially TGF-beta1, in neuron and astrocyte development. We will discuss some advances concerning the emerging scenario of TGF-beta1 and its signaling pathways as putative modulators of astrocyte biology and their implications as a novel mediator of cellular interactions in the CNS.
Collapse
Affiliation(s)
- Flávia Carvalho Alcantara Gomes
- Instituto de Ciências Biomédicas, Departamento de Anatomia, Universidade Federal do Rio de Janeiro, Centro de Ciências da Saúde, Bloco F, Ilha do Fundão, 21949-590 Rio de Janeiro, RJ, Brazil
| | | | | |
Collapse
|
24
|
Sulyok S, Wankell M, Alzheimer C, Werner S. Activin: an important regulator of wound repair, fibrosis, and neuroprotection. Mol Cell Endocrinol 2004; 225:127-32. [PMID: 15451577 DOI: 10.1016/j.mce.2004.07.011] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We recently identified the gene encoding the activin betaA chain as a novel injury-regulated gene. We showed that activin over-expression in the skin of transgenic mice enhances the speed of wound healing but also the scarring response. By contrast, inhibition of activin action by over-expression of the activin antagonist follistatin caused a severe delay in wound repair, but the quality of the healed wound was improved. In a search for activin-regulated genes in keratinocytes we identified the Mad1 transcription factor as a direct target of activin in these cells. Since Mad1 inhibits proliferation and induces differentiation of various cell types, our results suggest that activin regulates these processes in keratinocytes via induction of mad1. In addition to its role in the skin, we recently identified activin as a novel neuroprotective factor in vivo. Together with results from other laboratories, these findings suggest that activin is an important player in inflammation, repair and cytoprotection in various organs.
Collapse
Affiliation(s)
- Silke Sulyok
- Institute of Cell Biology, ETH Zürich, Hönggerberg, CH-8093 Zürich, Switzerland
| | | | | | | |
Collapse
|
25
|
Roussa E, Farkas LM, Krieglstein K. TGF-beta promotes survival on mesencephalic dopaminergic neurons in cooperation with Shh and FGF-8. Neurobiol Dis 2004; 16:300-10. [PMID: 15193287 DOI: 10.1016/j.nbd.2004.03.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2003] [Revised: 03/03/2004] [Accepted: 03/05/2004] [Indexed: 10/26/2022] Open
Abstract
Impaired neuronal survival is a key event in the development of degenerative diseases, such as Parkinson's disease (PD). Here we show that transforming growth factor beta (TGF-beta) acts directly on rat E14 midbrain dopaminergic neurons in vitro, its survival-promoting effect being not mediated by BDNF, NT-3, or GDNF. Treatment with TGF-beta, sonic hedgehog (Shh), or fibroblast growth factor-8 (FGF8) significantly increased number of tyrosine hydroxylase (TH)-immunoreactive neurons after 7 days, whereas application of these factors added together further increased number of TH-positive neurons, compared to single-factor treatments. Neutralization of endogenous TGF-beta, Shh, or FGF8 significantly reduced number of dopaminergic neurons. TGF-beta treatment decreased number of apoptotic cells, having no effect on cell proliferation. Neutralization of TGF-beta in vivo during chick E6-10 resulted in reduced number of midbrain dopaminergic neurons. The results suggest that TGF-beta is required for survival of mesencephalic dopaminergic neurons acting in cooperation with Shh and FGF8.
Collapse
Affiliation(s)
- Eleni Roussa
- Department for Neuroanatomy, Center of Anatomy, Georg-August-University Göttingen, 37075 Goettingen, Germany.
| | | | | |
Collapse
|
26
|
Sousa VDO, Romão L, Neto VM, Gomes FCA. Glial fibrillary acidic protein gene promoter is differently modulated by transforming growth factor-beta 1 in astrocytes from distinct brain regions. Eur J Neurosci 2004; 19:1721-30. [PMID: 15078546 DOI: 10.1111/j.1460-9568.2004.03249.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The expression of glial fibrillary acidic protein (GFAP), the major intermediate filament protein of mature astrocytes, is regulated under developmental and pathological conditions. Recently, we have investigated GFAP gene modulation by using a transgenic mouse bearing part of the GFAP gene promoter linked to the beta-galactosidase reporter gene. We demonstrated that cerebral cortex neurons activate the GFAP gene promoter, inducing transforming growth factor-beta 1 (TGF-beta 1) secretion by astrocytes. Here, we report that cortical neurons or conditioned medium derived from them do not activate the GFAP gene promoter of transgenic astrocytes derived from midbrain and cerebellum suggesting a neuroanatomical regional specificity of this phenomenon. Surprisingly, they do induce synthesis of TGF-beta 1 by these cells. Western blot and immunocytochemistry assays revealed wild distribution of TGF receptor in all subpopulations of astrocytes and expression of TGF-beta 1 in neurons derived from all regions, thus indicating that the unresponsiveness of the cerebellar and midbrain GFAP gene to TGF-beta 1 is not due to a defect in TGF-beta 1 signalling. Together, our data highlight the great complexity of neuron-glia interactions and might suggest a distinct mechanism underlying modulation of the GFAP gene in the heterogeneous population of astrocytes throughout the central nervous system.
Collapse
Affiliation(s)
- Vivian de Oliveira Sousa
- Departamento de Anatomia, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Centro de Ciências da Saúde, Bloco F, Ilha do Fundão, 21941-590, Rio de Janeiro, RJ, Brazil
| | | | | | | |
Collapse
|
27
|
Brionne TC, Tesseur I, Masliah E, Wyss-Coray T. Loss of TGF-beta 1 leads to increased neuronal cell death and microgliosis in mouse brain. Neuron 2004; 40:1133-45. [PMID: 14687548 DOI: 10.1016/s0896-6273(03)00766-9] [Citation(s) in RCA: 283] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
TGF-beta1 is a key regulator of diverse biological processes in many tissues and cell types, but its exact function in the developing and adult mammalian CNS is still unknown. We report that lack of TGF-beta1 expression in neonatal Tgfb1(-/-) mice results in a widespread increase in degenerating neurons accompanied by reduced expression of synaptophysin and laminin and a prominent microgliosis. Lack of TGF-beta1 also strongly reduces survival of primary neurons cultured from Tgfb1(-/-) mice. TGF-beta1 deficiency in adult Tgfb1(-/+) mice results in increased neuronal susceptibility to excitotoxic injury, whereas astroglial overexpression of TGF-beta1 protects adult mice against neurodegeneration in acute, excitotoxic and chronic injury paradigms. This study reveals a nonredundant function for TGF-beta1 in maintaining neuronal integrity and survival of CNS neurons and in regulating microglial activation. Because individual TGF-beta1 expression levels in the brain vary considerably between humans, this finding could have important implications for susceptibility to neurodegeneration.
Collapse
Affiliation(s)
- Thomas C Brionne
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | |
Collapse
|
28
|
Alzheimer C, Werner S. Fibroblast growth factors and neuroprotection. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2003; 513:335-51. [PMID: 12575827 DOI: 10.1007/978-1-4615-0123-7_12] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Several members of the FGF family, in particular FGF2, are intimately involved in neuronal protection and repair after ischemic, metabolic or traumatic brain injury. Expression of Fgf2 mRNA and protein is strongly upregulated after neuronal damage, with glial cells as the predominant source. Given its survival-promoting effects on cultured neurons, exogenous FGF2 was tested in several animal models of stroke and excitotoxic damage, in which it consistently proved protective against neuronal loss. FGF2 affords neuroprotection by interfering with a number of signaling pathways, including expression and gating of NMDA receptors, maintenance of Ca2+ homeostasis and regulation of ROS detoxifying enzymes. FGF2 prevents apoptosis by strengthening anti-apoptotic pathways and promotes neurogenesis in adult hippocampus after injury. The protective action of FGF2 has been linked to its augmenting effect on the lesion-induced upregulation of activin A, a member of the TGF-beta superfamily. Despite the well-documented benefits of FGF2 in animal models of stroke, there is currently no clinical development in stroke, after a phase II/III trial with FGF2 in acute stroke patients was discontinued because of an unfavorable risk-to-benefit ratio. As the molecular targets of FGF2 are going to be unraveled over the next years, new therapeutic strategies will hopefully emerge that enable us to influence the various protective mechanisms of FGF2 in a more specific fashion.
Collapse
Affiliation(s)
- Christian Alzheimer
- Institute of Physiology, University of Munich, Pettenkoferstr. 12, D-80336 Munich, Germany
| | | |
Collapse
|
29
|
Abstract
Activin is a member of the transforming growth factor beta family of growth and differentiation factors. Initially discovered as a protein that stimulates release of follicle-stimulating hormone, it is now well accepted as an important regulator of cell growth and differentiation. Most interestingly, a series of previous studies have revealed novel roles of activin in inflammation and repair. Our own results have provided evidence for an important function of activin in cutaneous wound repair as well as in neuroprotection, and these data will be summarized and discussed in this chapter.
Collapse
Affiliation(s)
- Miriam Wankell
- Institute of Cell Biology, ETH Zürich, Hönggerberg, CH-8093 Zürich, Switzerland
| | | | | | | |
Collapse
|
30
|
Rideout HJ, Dietrich P, Savalle M, Dauer WT, Stefanis L. Regulation of alpha-synuclein by bFGF in cultured ventral midbrain dopaminergic neurons. J Neurochem 2003; 84:803-13. [PMID: 12562524 DOI: 10.1046/j.1471-4159.2003.01574.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Alpha-synuclein is a neuronal protein that is implicated in the control of synaptic vesicle function and in Parkinson's disease (PD). Consequently, alterations of alpha-synuclein levels may play a role in neurotransmission and in PD pathogenesis. However, the factors that regulate alpha-synuclein levels are unknown. Growth factors mediate neurotrophic and plasticity effects in CNS neurons, and may play a role in disease states. Here we examine the regulation of alpha-synuclein levels in primary CNS neurons, with particular emphasis on dopaminergic neurons. E18 rat cortical neurons and dopaminergic neurons of E14 rat ventral midbrain showed an induction of alpha-synuclein protein levels with maturation in culture. Application of basic Fibroblast growth factor (bFGF) promoted alpha-synuclein expression selectively within dopaminergic, and not GABAergic or cortical neurons. This induction was blocked by actinomycin D, but not by inhibition of bFGF-induced glial proliferation. alpha-Synuclein levels were not altered by glial-derived neurotrophic factor (GDNF), or by apoptotic stimuli. We conclude that bFGF promotes alpha-synuclein expression in cultured ventral midbrain dopaminergic neurons through a direct transcriptional effect. These results suggest that distinct growth factors may thus mediate plasticity responses or influence disease states in ventral midbrain dopaminergic neurons.
Collapse
Affiliation(s)
- Hardy J Rideout
- Department of Neurology, Columbia University, Black Building Room 326, 650 W. 168th Street, New York, NY 10032, USA.
| | | | | | | | | |
Collapse
|
31
|
de Sampaio e Spohr TCL, Martinez R, da Silva EF, Neto VM, Gomes FCA. Neuro-glia interaction effects on GFAP gene: a novel role for transforming growth factor-beta1. Eur J Neurosci 2002; 16:2059-69. [PMID: 12473073 DOI: 10.1046/j.1460-9568.2002.02283.x] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Central nervous system (CNS) development is highly guided by microenvironment cues specially provided by neuron-glia interactions. By using a transgenic mouse bearing part of the gene promoter of the astrocytic maturation marker GFAP (glial fibrillary acidic protein) linked to the beta-galactosidase (beta-Gal) reporter gene, we previously demonstrated that cerebral cortical neurons increase transgenic beta-Gal astrocyte number and activate GFAP gene promoter by secretion of soluble factors in vitro. Here, we identified TGF-beta1 as the major mediator of this event. Identification of TGF-beta1 in neuronal and astrocyte extracts revealed that both cell types might synthesize this factor, however, addition of neurons to astrocyte monolayers greatly increased TGF-beta1 synthesis and secretion by astrocytes. Further, by exploiting the advantages of cell culture system we investigated the influence of neuron and astrocyte developmental stage on such interaction. We demonstrated that younger neurons derived from 14 embryonic days wild-type mice were more efficient in promoting astrocyte differentiation than those derived from 18 embryonic days mice. Similarly, astrocytes also exhibited timed-schedule developed responsiveness to neuronal influence with embryonic astrocytes being more responsive to neurons than newborn and late postnatal astrocytes. RT-PCR assays identified TGF-beta1 transcripts in young but not in old neurons, suggesting that inability to induce astrocyte differentiation is related to TGF-beta1 synthesis and secretion. Our work reveals an important role for neuron-glia interactions in astrocyte development and strongly implicates the involvement of TGF-beta1 in this event.
Collapse
|
32
|
Lenhard T, Schober A, Suter-Crazzolara C, Unsicker K. Fibroblast growth factor-2 requires glial-cell-line-derived neurotrophic factor for exerting its neuroprotective actions on glutamate-lesioned hippocampal neurons. Mol Cell Neurosci 2002; 20:181-97. [PMID: 12093153 DOI: 10.1006/mcne.2002.1134] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
FGF-2 is a potent neurotrophic factor for several populations of CNS neurons and has been shown to protect hippocampal neurons from glutamate-induced cell death in vitro and in vivo. Mechanisms underlying the neurotrophic and protective actions of FGF-2 have been resolved only in part. Using glutamate-treated cultured hippocampal neurons we show that FGF-2 shares its neuroprotective capacity with GDNF. Hippocampal neurons express glial-cell-line-derived neurotrophic factor (GDNF), its receptors c-Ret and the lipid-anchored GDNF family receptor-alpha1 (GFRalpha-1), and the FGF receptor 1 (FGFR I). Neutralizing antibodies to GDNF abolish the neuroprotective effect of FGF-2. In support of the notion that GDNF is required to permit the protective effects of FGF-2 we find that FGF-2 up-regulates GDNF and GFRalpha-1 in hippocampal neurons. Furthermore, FGF-2-induced GDNF causes enhanced phosphorylation of c-Ret and the signaling components Akt and Erk. A putative downstream target of FGF-2 and GDNF are bcl-2 gene family members, whose mRNAs are differentially up-regulated by the two factors. Together, these data suggest that GDNF is an important protective factor for glutamate-lesioned hippocampal neurons and an essential mediator of the neuroprotective actions of FGF-2.
Collapse
Affiliation(s)
- Thorsten Lenhard
- Department of Neuroanatomy and Interdisciplinary Center for Neurosciences (IZN), University of Heidelberg, Im Neuenheimer Feld 307, Germany
| | | | | | | |
Collapse
|
33
|
SiuYi Leung D, Unsicker K, Reuss B. Gap junctions modulate survival-promoting effects of fibroblast growth factor-2 on cultured midbrain dopaminergic neurons. Mol Cell Neurosci 2001; 18:44-55. [PMID: 11461152 DOI: 10.1006/mcne.2001.1002] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Fibroblast growth factor 2 (FGF-2) and glial cell line-derived neurotrophic factor (GDNF) support survival of dopaminergic midbrain neurons. Neurons are coupled by gap junctions, propagating metabolites and intracellular second messengers possibly mediating growth factor effects. We asked, therefore, whether gap junctions influence the survival-promoting effects of FGF-2 and GDNF. RT-PCR, Western blotting, and immunocytochemistry demonstrate that FGF-2 but not GDNF upregulates cx43 mRNA and immunoreactivity in rat embryonic day 14 midbrain cultures, whereas cx26, cx32, and cx45 were unchanged. In addition, functional coupling as assayed by the spread of neurobiotin was increased by FGF-2. Furthermore, the gap junction blocker oleamide abolished survival-promoting effects of FGF-2 on dopaminergic midbrain neurons. Together, these results support a direct role of gap junction communication for survival-promoting effects of FGF-2 on dopaminergic midbrain neurons, making gap junction communication a substantial parameter for neuron survival.
Collapse
Affiliation(s)
- D SiuYi Leung
- Neuroanatomy and Interdisciplinary Center for Neurosciences (IZN), University of Heidelberg, Im Neuenheimer Feld 307, Heidelberg, D-69120, Germany
| | | | | |
Collapse
|
34
|
Munz B, Tretter YP, Hertel M, Engelhardt F, Alzheimer C, Werner S. The roles of activins in repair processes of the skin and the brain. Mol Cell Endocrinol 2001; 180:169-77. [PMID: 11451588 DOI: 10.1016/s0303-7207(01)00514-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
A recent study from our laboratory demonstrated a strong upregulation of activin expression during cutaneous wound healing. To further analyze the role of activin A in skin morphogenesis and wound repair, we generated transgenic mice that overexpress activin A under the control of the keratin 14 promoter. The latter targets expression of transgenes to the basal, proliferating layer of the epidermis. Hetero- as well as homozygous transgenic animals were viable and fertile. However, they were smaller than non-transgenic littermates and they had smaller ears and shorter tails. Histological analysis of their skin revealed dermal hyperthickening, mainly due to the replacement of fatty tissue by connective tissue, and an increase in suprabasal, partially differentiated epidermal layers. After cutaneous injury, a strong enhancement of granulation tissue formation was observed. Furthermore, the extent of re-epithelialization was increased in some of the wounds. These data demonstrate that activin A is a potent stimulator of the wound healing process. Using an in vivo model of local brain injury, we found that activin A also plays a significant role in the early cellular response to neuronal damage. Expression of activin mRNA and protein is markedly upregulated within a few hours of injury. If applied exogenously, recombinant activin A is capable of rescuing neurons from acute cell death. Studying the interaction between bFGF, a well-established neuroprotective agent, which is currently being tested in stroke patients, and activin A, we arrived at the unexpected conclusion that it is the strong induction of activin A by bFGF which endows the latter with its beneficial actions in patients. These findings suggest that the development of substances directly targeting activin expression or receptor binding should offer new possibilities in the acute treatment of stroke and brain trauma.
Collapse
Affiliation(s)
- B Munz
- Max-Planck-Institute of Biochemistry, Am Klopferspitz 18a, D-82152, Martinsried, Germany
| | | | | | | | | | | |
Collapse
|
35
|
Pitts RL, Wang S, Jones EA, Symes AJ. Transforming growth factor-beta and ciliary neurotrophic factor synergistically induce vasoactive intestinal peptide gene expression through the cooperation of Smad, STAT, and AP-1 sites. J Biol Chem 2001; 276:19966-73. [PMID: 11259431 DOI: 10.1074/jbc.m011759200] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The cytokine ciliary neurotrophic factor (CNTF) and transforming growth factor-beta (TGF-beta) both induce transcription of the vasoactive intestinal peptide (VIP) gene through a 180-base pair cytokine response element (CyRE) in the VIP promoter. While CNTF induces STAT and AP-1 proteins to bind to cognate sites in the VIP CyRE, the mechanism through which TGF-beta acts to induce VIP gene transcription is not known. Here we show that Smad3 and Smad4 proteins can bind to two distinct sites within the VIP CyRE. These sites are absolutely required for the induction of VIP CyRE transcription by TGF-beta. TGF-beta induces endogenous Smad-containing complexes to bind to these sites in human neuroblastoma cells. CNTF and TGF-beta synergize to induce VIP mRNA expression and transcription through the VIP CyRE. This synergy is dependent on the Smad, STAT, and AP-1 sites, suggesting that these two independent cytokine pathways synergize through the cooperation of pathway-specific transcription factors binding to distinct sites within the VIP CyRE.
Collapse
Affiliation(s)
- R L Pitts
- Department of Pharmacology, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814, USA
| | | | | | | |
Collapse
|
36
|
Ovalle S, Zamanillo D, Andreu F, Farré AJ, Guitart X. Fibroblast growth factor-2 is selectively modulated in the rat brain by E-5842, a preferential sigma-1 receptor ligand and putative atypical antipsychotic. Eur J Neurosci 2001; 13:909-15. [PMID: 11264663 DOI: 10.1046/j.0953-816x.2001.01459.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Fibroblast growth factor-2 (FGF-2) is a member of a large family of trophic factors whose expression is regulated under several conditions in different areas of the brain. The goal of our experiments was to determine whether the administration of 4-(4-fluorophenyl)-1,2,3,6-tetrahydro-1-[4-(1,2,4-triazol-1-il)butyl] pyridine citrate (E-5842), a sigma-1 receptor ligand and putative atypical antipsychotic, could regulate the expression of FGF-2. After chronic treatment with E-5842 (21 days, and the animals killed 24 h after the last administration), an up-regulation was observed of the expression of FGF-2 mRNA in the prefrontal cortex and the striatum, and a down-regulation of the expression of FGF-2 mRNA in the hypothalamus of the rat brain. Acute treatment with E-5842 (one single administration and animals killed 6 h later) up-regulated FGF-2 expression in the prefrontal cortex, the striatum, the hypothalamus and the hippocampus in a dose-dependent manner. The acute up-regulation was transient and disappeared 24 h after E-5842 administration. The induction of FGF-2 in the striatum after repeated administration has been described for clozapine, but our data concerning regulation in the prefrontal cortex suggest that this effect is unique to E-5852 among other antipsychotics. Given the neuroprotective activity of FGF-2, the data presented here might be relevant to the deficit in cognition and other symptoms that appear in schizophrenia.
Collapse
Affiliation(s)
- S Ovalle
- Department of Neuropharmacology, Research Center, Laboratoris Esteve, S.A., Verge de Montserrat, 221, 08041-Barcelona, Spain
| | | | | | | | | |
Collapse
|
37
|
Reuss B, Unsicker K. Survival and differentiation of dopaminergic mesencephalic neurons are promoted by dopamine-mediated induction of FGF-2 in striatal astroglial cells. Mol Cell Neurosci 2000; 16:781-92. [PMID: 11124897 DOI: 10.1006/mcne.2000.0906] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Survival of dopaminergic (DAergic) midbrain neurons during development and after lesioning depends, in part, on the presence of astroglia-derived growth factors, as, e.g., fibroblast growth factor (FGF)-2. Astrocytes express DA receptors in a brain-region-specific manner. We show here that DA (10(-3) to 10(-6) mol/liter) applied continuously for 12 h or as a 10-min pulse significantly upregulates FGF-2 immunoreactivity quantified by Western blot and densitometry in astrocytes cultured from two target areas of DAergic neurons, striatum and cortex, but not in mesencephalic astroglia. Semiquantitative competitive RT-PCR confirmed the increase in FGF-2 on the mRNA level. The effects were specific in that glutamate, which can also activate receptors on astroglial cells, did not influence FGF-2 synthesis. In addition to the DA-mediated increase in FGF-2 synthesis the capability of conditioned medium (CM) from DA-stimulated striatal and cortical astrocytes to promote survival and process formation of cultured rat DAergic neurons was significantly enhanced. These effects could be fully blocked by preincubation of the CM with an FGF-2-specific polyclonal antiserum. Our results suggest that DA released from DAergic axon terminals in target regions of DAergic neurons and astroglial FGF-2 production are interdependent in that DA triggers synthesis of FGF-2, which, in turn enhances survival and differentiation of DAergic neurons.
Collapse
Affiliation(s)
- B Reuss
- Neuroanatomy and Interdisciplinary Center for Neurosciences (IZN), University of Heidelberg, Im Neuenheimer Feld 307, Heidelberg, D-69120, Germany.
| | | |
Collapse
|
38
|
Flores C, Stewart J. Changes in astrocytic basic fibroblast growth factor expression during and after prolonged exposure to escalating doses of amphetamine. Neuroscience 2000; 98:287-93. [PMID: 10854759 DOI: 10.1016/s0306-4522(00)00115-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We have shown that brief exposure to amphetamine leads to sustained glutamate-dependent increases in expression of the neurotrophic, neuroprotective factor, basic fibroblast growth factor, in astrocytes in dopaminergic cell body regions and that blockade of basic fibroblast growth factor in this region prevents the development of behavioral sensitization to amphetamine. Here we examine the effects of prolonged exposure to an escalating-dose regimen of amphetamine known to induce long-lasting sensitization to amphetamine and leading to increases in neuronal dendritic length and spine density in nucleus accumbens and prefrontal cortex and to decreases in spine density in occipital cortex. Astrocytic basic fibroblast growth factor immunoreactivity was increased in both dopaminergic cell body and terminal regions one week after termination of a two-week amphetamine treatment (1-4mg/kg). These effects were not evident one week after a five-week treatment (1-9mg/kg) and, in fact, one month later basic fibroblast growth factor levels in cell body regions were decreased. In the occipital cortex, basic fibroblast growth factor immunoreactivity was decreased one week after the two-week amphetamine treatment, but was not different from that seen in saline-treated animals after the five-week treatment. Increased astrocytic basic fibroblast growth factor expression appears to be an early, but relatively prolonged, response to amphetamine exposure and seems to parallel structural changes induced by repeated drug exposure.These findings suggest that basic fibroblast growth factor may participate in the development of structural changes brought about by amphetamine. The fact that the basic fibroblast growth factor response is not maintained after prolonged intense exposure to amphetamine suggests that the factors that initially induce basic fibroblast growth factor expression are self-regulating.
Collapse
Affiliation(s)
- C Flores
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Quebec, Montreal, Canada, H3G 1M8
| | | |
Collapse
|
39
|
Tretter YP, Hertel M, Munz B, ten Bruggencate G, Werner S, Alzheimer C. Induction of activin A is essential for the neuroprotective action of basic fibroblast growth factor in vivo. Nat Med 2000; 6:812-5. [PMID: 10888932 DOI: 10.1038/77548] [Citation(s) in RCA: 125] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Exogenous application of neurotrophic growth factors has emerged as a new and particularly promising approach not only to promote functional recovery after acute brain injury but also to protect neurons against the immediate effect of the injury. Among the various growth factors and cytokines studied so far, the neuroprotective and neurotrophic profile of basic fibroblast growth factor (bFGF) is the best documented. Using an animal model of acute excitotoxic brain injury, we report here that the neuroprotective action of bFGF, which is now being tested in stroke patients, depends on the induction of activin A, a member of the transforming growth factor-beta superfamily. Our evidence for this previously unknown mechanism of action of bFGF is that bFGF strongly enhanced lesion-associated induction of activin A; in the presence of the activin-neutralizing protein follistatin, bFGF was no longer capable of rescuing neurons from excitotoxic death; and recombinant activin A exerted a neuroprotective effect by itself. Our data indicate that the development of substances influencing activin expression or receptor binding should offer new ways to fight neuronal loss in ischemic and traumatic brain injury.
Collapse
Affiliation(s)
- Y P Tretter
- Department of Physiology, University of Munich, Pettenkoferstr.12, D-80336 München, Germany
| | | | | | | | | | | |
Collapse
|
40
|
Pataky DM, Borisoff JF, Fernandes KJ, Tetzlaff W, Steeves JD. Fibroblast growth factor treatment produces differential effects on survival and neurite outgrowth from identified bulbospinal neurons in vitro. Exp Neurol 2000; 163:357-72. [PMID: 10833309 DOI: 10.1006/exnr.2000.7365] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The in vivo application of appropriate trophic factors may enhance regeneration of bulbospinal projections after spinal cord injury. Currently, little is known about the sensitivities of specific bulbospinal neuron populations to the many identified trophic factors. We devised novel in vitro assays to study trophic effects on the survival and neurite outgrowth of identified bulbospinal neurons. Carbocyanine dye crystals implanted into the cervical spinal cord of embryonic day (E)5 chick embryos retrogradely labeled developing bulbospinal neurons. On E8, dissociated cultures containing labeled bulbospinal neurons were prepared. Fibroblast growth factor (FGF)-2 (but not FGF-1) promoted the survival of bulbospinal neurons. FGF receptor expression was widespread in the E8 brainstem, but not detected in young bulbospinal neurons, suggesting that nonneuronal cells mediated the FGF-stimulated survival response. Astrocytes synthesize a variety of trophic factors, and astrocyte-conditioned medium (ACM) also promoted the survival of bulbospinal neurons. As might be expected, FGF-2 function blocking antibodies did not suppress ACM-promoted survival, nor did an ELISA detect FGF-2 in ACM. This suggests that nonneuronal cells synthesize other factors in response to exogenous FGF-2 which promote the survival of bulbospinal neurons. Focusing on vestibulospinal neurons, dissociated (survival assay) or explant (neurite outgrowth assay) cultures were prepared. FGF-2 promoted both survival and neurite outgrowth of identified vestibulospinal neurons. Interestingly, FGF-1 promoted neurite outgrowth but not survival; the converse was true of FGF-9. Thus, differential effects of specific growth factors on survival or neurite outgrowth of bulbospinal neurons were distinguished.
Collapse
Affiliation(s)
- D M Pataky
- CORD, Collaboration On Repair Discoveries, University of British Columbia, 6270 University Boulevard, Vancouver, British Columbia, V6T 1Z4, Canada
| | | | | | | | | |
Collapse
|
41
|
Deigner HP, Haberkorn U, Kinscherf R. Apoptosis modulators in the therapy of neurodegenerative diseases. Expert Opin Investig Drugs 2000; 9:747-64. [PMID: 11060707 DOI: 10.1517/13543784.9.4.747] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Apoptosis is a prerequisite to model the developing nervous system. However, an increased rate of cell death in the adult nervous system underlies neurodegenerative disease and is a hallmark of multiple sclerosis (MS) Alzheimer's- (AD), Parkinson- (PD), or Huntington's disease (HD). Cell surface receptors (e.g., CD95/APO-1/Fas; TNF receptor) and their ligands (CD95-L; TNF) as well as evolutionarily conserved mechanisms involving proteases, mitochondrial factors (e.g. , Bcl-2-related proteins, reactive oxygen species, mitochondrial membrane potential, opening of the permeability transition pore) or p53 participate in the modulation and execution of cell death. Effectors comprise oxidative stress, inflammatory processes, calcium toxicity and survival factor deficiency. Therapeutic agents are being developed to interfere with these events, thus conferring the potential to be neuroprotective. In this context, drugs with anti-oxidative properties, e.g., flupirtine, N-acetylcysteine, idebenone, melatonin, but also novel dopamine agonists (ropinirole and pramipexole) have been shown to protect neuronal cells from apoptosis and thus have been suggested for treating neurodegenerative disorders like AD or PD. Other agents like non-steroidal anti-inflammatory drugs (NSAIDs) partly inhibit cyclooxygenase (COX) expression, as well as having a positive influence on the clinical expression of AD. Distinct cytokines, growth factors and related drug candidates, e.g., nerve growth factor (NGF), or members of the transforming growth factor-beta (TGF-beta ) superfamily, like growth and differentiation factor 5 (GDF-5), are shown to protect tyrosine hydroxylase or dopaminergic neurones from apoptosis. Furthermore, peptidergic cerebrolysin has been found to support the survival of neurones in vitro and in vivo. Treatment with protease inhibitors are suggested as potential targets to prevent DNA fragmentation in dopaminergic neurones of PD patients. Finally, CRIB (cellular replacement by immunoisolatory biocapsule) is an auspicious gene therapeutical approach for human NGF secretion, which has been shown to protect cholinergic neurones from cell death when implanted in the brain. This review summarises and evaluates novel aspects of anti-apoptotic concepts and pharmacological intervention including gene therapeutical approaches currently being proposed or utilised to treat neurodegenerative diseases.
Collapse
Affiliation(s)
- H P Deigner
- Anatomy and Cell Biology III University of Heidelberg, Germany
| | | | | |
Collapse
|
42
|
Abstract
Dopamine (DA) is a monoamine neurotransmitter of both central and peripheral nervous system. Its role in the neural-immune communication has been discussed in the present review. Results reveal that in vivo damage or stimulation of specific central dopaminergic system suppresses or enhances functional activities of the immune effector cells. The possible influences of other immunomodulators of the brain by altering brain DA may be the underlying mechanism. Direct effects of DA on the immune effector cells are also contradictory, it is suppressive in vitro, while in pharmacological doses, it is mostly stimulatory in vivo. The possible mechanisms have been discussed. Lastly, future areas of relevance on DA and immunity have been highlighted to advance our knowledge regarding DA as an immune regulator.
Collapse
Affiliation(s)
- S Basu
- Department of Medical Oncology, Chittaranjan National Cancer Institute, Calcutta, India
| | | |
Collapse
|