1
|
Lopez-Rodriguez D, Rohrbach A, Lanzillo M, Gervais M, Croizier S, Langlet F. Ontogeny of ependymoglial cells lining the third ventricle in mice. Front Endocrinol (Lausanne) 2023; 13:1073759. [PMID: 36686420 PMCID: PMC9849764 DOI: 10.3389/fendo.2022.1073759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 12/02/2022] [Indexed: 01/07/2023] Open
Abstract
Introduction During hypothalamic development, the germinative neuroepithelium gives birth to diverse neural cells that regulate numerous physiological functions in adulthood. Methods Here, we studied the ontogeny of ependymal cells in the mouse mediobasal hypothalamus using the BrdU approach and publicly available single-cell RNAseq datasets. Results We observed that while typical ependymal cells are mainly produced at E13, tanycyte birth depends on time and subtypes and lasts up to P8. Typical ependymocytes and β tanycytes are the first to arise at the top and bottom of the dorsoventral axis around E13, whereas α tanycytes emerge later in development, generating an outside-in dorsoventral gradient along the third ventricle. Additionally, α tanycyte generation displayed a rostral-to-caudal pattern. Finally, tanycytes mature progressively until they reach transcriptional maturity between P4 and P14. Discussion Altogether, this data shows that ependyma generation differs in time and distribution, highlighting the heterogeneity of the third ventricle.
Collapse
Affiliation(s)
- David Lopez-Rodriguez
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Antoine Rohrbach
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Marc Lanzillo
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Manon Gervais
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Sophie Croizier
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Fanny Langlet
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
2
|
Prakash N. Developmental pathways linked to the vulnerability of adult midbrain dopaminergic neurons to neurodegeneration. Front Mol Neurosci 2022; 15:1071731. [PMID: 36618829 PMCID: PMC9815185 DOI: 10.3389/fnmol.2022.1071731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
The degeneration of dopaminergic and other neurons in the aging brain is considered a process starting well beyond the infantile and juvenile period. In contrast to other dopamine-associated neuropsychiatric disorders, such as schizophrenia and drug addiction, typically diagnosed during adolescence or young adulthood and, thus, thought to be rooted in the developing brain, Parkinson's Disease (PD) is rarely viewed as such. However, evidences have accumulated suggesting that several factors might contribute to an increased vulnerability to death of the dopaminergic neurons at an already very early (developmental) phase in life. Despite the remarkable ability of the brain to compensate such dopamine deficits, the early loss or dysfunction of these neurons might predispose an individual to suffer from PD because the critical threshold of dopamine function will be reached much earlier in life, even if the time-course and strength of naturally occurring and age-dependent dopaminergic cell death is not markedly altered in this individual. Several signaling and transcriptional pathways required for the proper embryonic development of the midbrain dopaminergic neurons, which are the most affected in PD, either continue to be active in the adult mammalian midbrain or are reactivated at the transition to adulthood and under neurotoxic conditions. The persistent activity of these pathways often has neuroprotective functions in adult midbrain dopaminergic neurons, whereas the reactivation of silenced pathways under pathological conditions can promote the survival and even regeneration of these neurons in the lesioned or aging brain. This article summarizes our current knowledge about signaling and transcription factors involved in midbrain dopaminergic neuron development, whose reduced gene dosage or signaling activity are implicated in a lower survival rate of these neurons in the postnatal or aging brain. It also discusses the evidences supporting the neuroprotection of the midbrain dopaminergic system after the external supply or ectopic expression of some of these secreted and nuclear factors in the adult and aging brain. Altogether, the timely monitoring and/or correction of these signaling and transcriptional pathways might be a promising approach to a much earlier diagnosis and/or prevention of PD.
Collapse
|
3
|
Abstract
A hypercaloric diet combined with a sedentary lifestyle is a major risk factor for the development of insulin resistance, type 2 diabetes mellitus (T2DM) and associated comorbidities. Standard treatment for T2DM begins with lifestyle modification, and includes oral medications and insulin therapy to compensate for progressive β-cell failure. However, current pharmaceutical options for T2DM are limited in that they do not maintain stable, durable glucose control without the need for treatment intensification. Furthermore, each medication is associated with adverse effects, which range from hypoglycaemia to weight gain or bone loss. Unexpectedly, fibroblast growth factor 1 (FGF1) and its low mitogenic variants have emerged as potentially safe candidates for restoring euglycaemia, without causing overt adverse effects. In particular, a single peripheral injection of FGF1 can lower glucose to normal levels within hours, without the risk of hypoglycaemia. Similarly, a single intracerebroventricular injection of FGF1 can induce long-lasting remission of the diabetic phenotype. This Review discusses potential mechanisms by which centrally administered FGF1 improves central glucose-sensing and peripheral glucose uptake in a sustained manner. Specifically, we explore the potential crosstalk between FGF1 and glucose-sensing neuronal circuits, hypothalamic neural stem cells and synaptic plasticity. Finally, we highlight therapeutic considerations of FGF1 and compare its metabolic actions with FGF15 (rodents), FGF19 (humans) and FGF21.
Collapse
Affiliation(s)
- Emanuel Gasser
- Gene Expression Laboratory, Salk Institute for Biological Studies
| | - Christopher P Moutos
- Gene Expression Laboratory, Salk Institute for Biological Studies
- Howard Hughes Medical Institute, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California 92037, USA
- College of Medicine, University of Arkansas for Medical Sciences, 4301 West Markham Street, Little Rock, Arkansas 72205, USA
| | - Michael Downes
- Gene Expression Laboratory, Salk Institute for Biological Studies
| | - Ronald M Evans
- Gene Expression Laboratory, Salk Institute for Biological Studies
- Howard Hughes Medical Institute, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California 92037, USA
| |
Collapse
|
4
|
Makani V, Jang YG, Christopher K, Judy W, Eckstein J, Hensley K, Chiaia N, Kim DS, Park J. BBB-Permeable, Neuroprotective, and Neurotrophic Polysaccharide, Midi-GAGR. PLoS One 2016; 11:e0149715. [PMID: 26939023 PMCID: PMC4777489 DOI: 10.1371/journal.pone.0149715] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 02/04/2016] [Indexed: 12/19/2022] Open
Abstract
An enormous amount of efforts have been poured to find an effective therapeutic agent for the treatment of neurodegenerative diseases including Alzheimer’s disease (AD). Among those, neurotrophic peptides that regenerate neuronal structures and increase neuron survival show a promise in slowing neurodegeneration. However, the short plasma half-life and poor blood-brain-barrier (BBB)-permeability of neurotrophic peptides limit their in vivo efficacy. Thus, an alternative neurotrophic agent that has longer plasma half-life and better BBB-permeability has been sought for. Based on the recent findings of neuroprotective polysaccharides, we searched for a BBB-permeable neuroprotective polysaccharide among natural polysaccharides that are approved for human use. Then, we discovered midi-GAGR, a BBB-permeable, long plasma half-life, strong neuroprotective and neurotrophic polysaccharide. Midi-GAGR is a 4.7kD cleavage product of low acyl gellan gum that is approved by FDA for human use. Midi-GAGR protected rodent cortical neurons not only from the pathological concentrations of co-/post-treated free reactive radicals and Aβ42 peptide but also from activated microglial cells. Moreover, midi-GAGR showed a good neurotrophic effect; it enhanced neurite outgrowth and increased phosphorylated cAMP-responsive element binding protein (pCREB) in the nuclei of primary cortical neurons. Furthermore, intra-nasally administered midi-GAGR penetrated the BBB and exerted its neurotrophic effect inside the brain for 24 h after one-time administration. Midi-GAGR appears to activate fibroblast growth factor receptor 1 (FGFR1) and its downstream neurotrophic signaling pathway for neuroprotection and CREB activation. Additionally, 14-day intranasal administration of midi-GAGR not only increased neuronal activity markers but also decreased hyperphosphorylated tau, a precursor of neurofibrillary tangle, in the brains of the AD mouse model, 3xTg-AD. Taken together, midi-GAGR with good BBB-permeability, long plasma half-life, and strong neuroprotective and neurotrophic effects has a great therapeutic potential for the treatment of neurodegenerative diseases, especially AD.
Collapse
Affiliation(s)
- Vishruti Makani
- Department of Neurosciences, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio, United States of America
| | - Yong-gil Jang
- Department of Neurosciences, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio, United States of America
| | - Kevin Christopher
- Department of Neurosciences, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio, United States of America
| | - Wesley Judy
- Department of Neurosciences, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio, United States of America
| | - Jacob Eckstein
- Department of Neurosciences, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio, United States of America
| | - Kenneth Hensley
- Department of Pathology, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio, United States of America
| | - Nicolas Chiaia
- Department of Neurosciences, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio, United States of America
| | - Dong-Shik Kim
- Department of Chemical Engineering, College of Engineering, University of Toledo, Toledo, Ohio, United States of America
| | - Joshua Park
- Department of Neurosciences, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio, United States of America
- * E-mail:
| |
Collapse
|
5
|
Morton GJ, Matsen ME, Bracy DP, Meek TH, Nguyen HT, Stefanovski D, Bergman RN, Wasserman DH, Schwartz MW. FGF19 action in the brain induces insulin-independent glucose lowering. J Clin Invest 2013; 123:4799-808. [PMID: 24084738 PMCID: PMC3809800 DOI: 10.1172/jci70710] [Citation(s) in RCA: 168] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 08/01/2013] [Indexed: 12/17/2022] Open
Abstract
Insulin-independent glucose disposal (referred to as glucose effectiveness [GE]) is crucial for glucose homeostasis and, until recently, was thought to be invariable. However, GE is reduced in type 2 diabetes and markedly decreased in leptin-deficient ob/ob mice. Strategies aimed at increasing GE should therefore be capable of improving glucose tolerance in these animals. The gut-derived hormone FGF19 has previously been shown to exert potent antidiabetic effects in ob/ob mice. In ob/ob mice, we found that systemic FGF19 administration improved glucose tolerance through its action in the brain and that a single, low-dose i.c.v. injection of FGF19 dramatically improved glucose intolerance within 2 hours. Minimal model analysis of glucose and insulin data obtained during a frequently sampled i.v. glucose tolerance test showed that the antidiabetic effect of i.c.v. FGF19 was solely due to increased GE and not to changes of either insulin secretion or insulin sensitivity. The mechanism underlying this effect appears to involve increased metabolism of glucose to lactate. Together, these findings implicate the brain in the antidiabetic action of systemic FGF19 and establish the brain’s capacity to rapidly, potently, and selectively increase insulin-independent glucose disposal.
Collapse
Affiliation(s)
- Gregory J. Morton
- Diabetes and Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, Washington, USA.
Department of Molecular Physiology and Biophysics, Vanderbilt School of Medicine, Nashville, Tennessee, USA.
Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Miles E. Matsen
- Diabetes and Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, Washington, USA.
Department of Molecular Physiology and Biophysics, Vanderbilt School of Medicine, Nashville, Tennessee, USA.
Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Deanna P. Bracy
- Diabetes and Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, Washington, USA.
Department of Molecular Physiology and Biophysics, Vanderbilt School of Medicine, Nashville, Tennessee, USA.
Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Thomas H. Meek
- Diabetes and Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, Washington, USA.
Department of Molecular Physiology and Biophysics, Vanderbilt School of Medicine, Nashville, Tennessee, USA.
Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Hong T. Nguyen
- Diabetes and Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, Washington, USA.
Department of Molecular Physiology and Biophysics, Vanderbilt School of Medicine, Nashville, Tennessee, USA.
Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Darko Stefanovski
- Diabetes and Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, Washington, USA.
Department of Molecular Physiology and Biophysics, Vanderbilt School of Medicine, Nashville, Tennessee, USA.
Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Richard N. Bergman
- Diabetes and Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, Washington, USA.
Department of Molecular Physiology and Biophysics, Vanderbilt School of Medicine, Nashville, Tennessee, USA.
Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - David H. Wasserman
- Diabetes and Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, Washington, USA.
Department of Molecular Physiology and Biophysics, Vanderbilt School of Medicine, Nashville, Tennessee, USA.
Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Michael W. Schwartz
- Diabetes and Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, Washington, USA.
Department of Molecular Physiology and Biophysics, Vanderbilt School of Medicine, Nashville, Tennessee, USA.
Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
6
|
Ryan KK, Kohli R, Gutierrez-Aguilar R, Gaitonde SG, Woods SC, Seeley RJ. Fibroblast growth factor-19 action in the brain reduces food intake and body weight and improves glucose tolerance in male rats. Endocrinology 2013; 154. [PMID: 23183168 PMCID: PMC3529386 DOI: 10.1210/en.2012-1891] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Fibroblast growth factor-19 (FGF19) and its rodent ortholog, FGF15, are hormones produced in the distal small intestine and secreted into the circulation after a meal. In addition to controlling the enterohepatic circulation of bile acids, FGF15/19 also regulates systemic lipid and glucose metabolism. In these experiments we investigated the hypothesis that, like other gut-derived postprandial hormones, FGF15/19 can act in the central nervous system to elicit its metabolic effects. We found that FGF-receptors 1 and 4 are present in rat hypothalamus, and that their expression was reduced by up to 60% in high-fat fed rats relative to lean controls. Consistent with a potential role for brain FGF15/19 signaling to regulate energy and glucose homeostasis, and with a previous report that intracerebroventricular (i.c.v.) administration of FGF19 increases energy expenditure, we report that acute i.c.v. FGF19 reduces 24-h food intake and body weight, and acutely improves glucose tolerance. Conversely, i.c.v. administration of an FGF-receptor inhibitor increases food intake and impairs glucose tolerance, suggesting a physiological role for brain FGF receptor signaling. Together, these findings identify the central nervous system as a potentially important target for the beneficial effects of FGF19 in the treatment of obesity and diabetes.
Collapse
Affiliation(s)
- Karen K Ryan
- The University of Cincinnati College of Medicine, Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, Cincinnati, OH 45237, USA.
| | | | | | | | | | | |
Collapse
|
7
|
Müller TD, Sullivan LM, Habegger K, Yi CX, Kabra D, Grant E, Ottaway N, Krishna R, Holland J, Hembree J, Perez-Tilve D, Pfluger PT, DeGuzman MJ, Siladi ME, Kraynov VS, Axelrod DW, DiMarchi R, Pinkstaff JK, Tschöp MH. Restoration of leptin responsiveness in diet-induced obese mice using an optimized leptin analog in combination with exendin-4 or FGF21. J Pept Sci 2012; 18:383-93. [PMID: 22565812 DOI: 10.1002/psc.2408] [Citation(s) in RCA: 122] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2012] [Revised: 03/02/2012] [Accepted: 03/05/2012] [Indexed: 11/07/2022]
Abstract
The identification of leptin as a mediator of body weight regulation provided much initial excitement for the treatment of obesity. Unfortunately, leptin monotherapy is insufficient in reversing obesity in rodents or humans. Recent findings suggest that amylin is able to restore leptin sensitivity and when used in combination with leptin enhances body weight loss in obese rodents and humans. However, as the uniqueness of this combination therapy remains unclear, we assessed whether co-administration of leptin with other weight loss-inducing hormones equally restores leptin responsiveness in diet-induced obese (DIO) mice. Accordingly, we report here the design and characterization of a series of site-specifically enhanced leptin analogs of high potency and sustained action that, when administered in combination with exendin-4 or fibroblast growth factor 21 (FGF21), restores leptin responsiveness in DIO mice after an initial body weight loss of 30%. Using either combination, body weight loss was enhanced compared with either exendin-4 or FGF21 monotherapy, and leptin alone was sufficient to maintain the reduced body weight. In contrast, leptin monotherapy proved ineffective when identical weight loss was induced by caloric restriction alone over a comparable time. Accordingly, we find that a hypothalamic counter-regulatory response to weight loss, assessed using changes in hypothalamic agouti related peptide (AgRP) levels, is triggered by caloric restriction, but blunted by treatment with exendin-4. We conclude that leptin re-sensitization requires pharmacotherapy but does not appear to be restricted to a unique signaling pathway. Our findings provide preclinical evidence that high activity, long-acting leptin analogs are additively efficacious when used in combination with other weight-lowering agents.
Collapse
Affiliation(s)
- Timo D Müller
- Metabolic Disease Institute, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45230, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Iwata K, Kinoshita M, Yamada S, Imamura T, Uenoyama Y, Tsukamura H, Maeda KI. Involvement of brain ketone bodies and the noradrenergic pathway in diabetic hyperphagia in rats. J Physiol Sci 2011; 61:103-13. [PMID: 21234734 PMCID: PMC10717331 DOI: 10.1007/s12576-010-0127-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2010] [Accepted: 12/05/2010] [Indexed: 10/18/2022]
Abstract
Uncontrolled type 1 diabetes leads to hyperphagia and severe ketosis. This study was conducted to test the hypothesis that ketone bodies act on the hindbrain as a starvation signal to induce diabetic hyperphagia. Injection of an inhibitor of monocarboxylate transporter 1, a ketone body transporter, into the fourth ventricle normalized the increase in food intake in streptozotocin (STZ)-induced diabetic rats. Blockade of catecholamine synthesis in the hypothalamic paraventricular nucleus (PVN) also restored food intake to normal levels in diabetic animals. On the other hand, hindbrain injection of the ketone body induced feeding, hyperglycemia, and fatty acid mobilization via increased sympathetic activity and also norepinephrine release in the PVN. This result provides evidence that hyperphagia in STZ-induced type 1 diabetes is signaled by a ketone body sensed in the hindbrain, and mediated by noradrenergic inputs to the PVN.
Collapse
Affiliation(s)
- Kinuyo Iwata
- Laboratory for Reproductive Science, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8601 Japan
| | - Mika Kinoshita
- Laboratory for Reproductive Science, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8601 Japan
| | - Shunji Yamada
- Laboratory for Reproductive Science, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8601 Japan
| | - Takuya Imamura
- Laboratory for Biodiversity, Global COE Program, Graduate School of Science, Kyoto University, Kyoto, 606-8502 Japan
| | - Yoshihisa Uenoyama
- Laboratory for Reproductive Science, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8601 Japan
| | - Hiroko Tsukamura
- Laboratory for Reproductive Science, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8601 Japan
| | - Kei-ichiro Maeda
- Laboratory for Reproductive Science, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8601 Japan
| |
Collapse
|
9
|
Topp S, Stigloher C, Komisarczuk AZ, Adolf B, Becker TS, Bally-Cuif L. Fgf signaling in the zebrafish adult brain: Association of Fgf activity with ventricular zones but not cell proliferation. J Comp Neurol 2008; 510:422-39. [DOI: 10.1002/cne.21802] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
10
|
Mathew TC. Regional analysis of the ependyma of the third ventricle of rat by light and electron microscopy. Anat Histol Embryol 2008; 37:9-18. [PMID: 18197894 DOI: 10.1111/j.1439-0264.2007.00786.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Ependymal lining of cerebral ventricles lies at the interface between the ventricular cavities and the brain parenchyma. Ependymal cells are involved in various functions within the brain and play a major role in the production of the chemical principals of the cerebrospinal fluid. Histological studies on the regional variation of the third ventricular ependyma and the subependyma of adult rats were carried out by light and electron microscopic methods. For light microscopic analysis, methacrylate sections were used. In addition to the routine haematoxylin and eosin (H and E) staining for histological studies, the sections were stained with toluidine blue, cresyl violet and periodic acid Schiff's reagent (PAS). A regional analysis of the ependyma of the third ventricle showed that in most regions the ependyma was monolayered. The sidewalls and floor of the ventral portion of the third ventricle showed a multilayered ependyma. For descriptive purposes at the light microscopic level, the ependymal cells were classified, based on the cell shape (flat, cuboidal or columnar), presence or absence of cilia and the number of cytoplasmic granules present in the cells. Studies of transmission electron microscope have shown that these granules represent the cell organelles of the ependyma. The subependyma also showed a regional morphological variation, and, in most instances, contained glial and neuronal elements. In regions of specific brain nuclei, neurons were the major cell type of the subependyma. PAS staining did not show any positive granules in the ependymal cytosol. Characteristic supraependymal elements were present at the ependymal surface of the third ventricle.
Collapse
Affiliation(s)
- T C Mathew
- Department of Medical Laboratory Sciences, Faculty of Allied Health Sciences, Kuwait University, Kuwait.
| |
Collapse
|
11
|
Zhao M, Li D, Shimazu K, Zhou YX, Lu B, Deng CX. Fibroblast growth factor receptor-1 is required for long-term potentiation, memory consolidation, and neurogenesis. Biol Psychiatry 2007; 62:381-90. [PMID: 17239352 DOI: 10.1016/j.biopsych.2006.10.019] [Citation(s) in RCA: 123] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2006] [Revised: 09/13/2006] [Accepted: 10/11/2006] [Indexed: 10/23/2022]
Abstract
BACKGROUND Although substantial evidence supports the view that adult neurogenesis is involved in learning and memory, how newly generated neurons contribute to the cognitive process remains unknown. Fibroblast growth factor 2 (FGF-2) is known to stimulate the proliferation of neuronal progenitor cells (NPCs) in adult brain. Using conditional knockout mice that lack brain expression of FGFR1, a major receptor for FGF-2, we have investigated the role of adult neurogenesis in hippocampal synaptic plasticity and learning and memory. METHODS The Fgfr1 conditional knockout mice were generated by crossing the Fgfr1-null line, the Fgfr1-flox line, and the Nestin-Cre transgenic mice. Bromodeoxyuridine (BrdU) labeling, slice electrophysiology, and Morris Water Maze experiments were performed with the Fgfr1 conditional mutant mice. RESULTS Bromodeoxyuridine labeling experiments demonstrate that FGFR1 is required for the proliferation of NPCs as well as generation of new neurons in the adult dentate gyrus (DG). Moreover, deficits in neurogenesis in Fgfr1 mutant mice are accompanied by a severe impairment of long-term potentiation (LTP) at the medial perforant path (MPP)-granule neuron synapses in the hippocampal dentate. Moreover, the Fgfr1 mutant mice exhibit significant deficits in memory consolidation but not spatial learning. CONCLUSIONS Our study suggests a critical role of FGFR1 in adult neurogenesis in vivo, provides a potential link between proliferative neurogenesis and dentate LTP, and raises the possibility that adult neurogenesis might contribute to memory consolidation.
Collapse
Affiliation(s)
- Mingrui Zhao
- Section on Neural Development and Plasticity, Laboratory of Cellular & Synaptic Neurophysiology, National Institute of Child Health and Human Development, Bethesda, Maryland 20892, USA
| | | | | | | | | | | |
Collapse
|
12
|
Sun HD, Malabunga M, Tonra JR, DiRenzo R, Carrick FE, Zheng H, Berthoud HR, McGuinness OP, Shen J, Bohlen P, Leibel RL, Kussie P. Monoclonal antibody antagonists of hypothalamic FGFR1 cause potent but reversible hypophagia and weight loss in rodents and monkeys. Am J Physiol Endocrinol Metab 2007; 292:E964-76. [PMID: 17132826 DOI: 10.1152/ajpendo.00089.2006] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
We generated three fully human monoclonal antibody antagonists against fibroblast growth factor receptor-1 (FGFR1) that potently block FGF signaling. We found that antibodies targeting the c-splice form of the receptor (FGFR1c) were anorexigenic when administered intraperitoneally three times weekly to mice, resulting in rapid, dose-dependent weight loss that plateaued (for doses>4 mg/kg) at 35-40% in 2 wk. Animals appeared healthy during treatment and regained their normal body weights and growth trajectories upon clearance of the antibodies from the bloodstream. Measurements of food consumption and energy expenditure indicated that the rapid weight loss was induced primarily by decreased energy intake and not by increased energy expenditure or cachexia and was accompanied by a greater reduction in fat than lean body mass. Hypophagia was not caused through malaise or illness, as indicated by absence of conditioned taste aversion, pica behavior, and decreased need-induced salt intake in rats. In support of a hypothalamic site of action, we found that, after intraperitoneal injections, anti-FGFR1c (IMC-A1), but not a control antibody, accumulated in the median eminence and adjacent mediobasal hypothalamus and that FGFR1c is enriched in the hypothalamus of mice. Furthermore, a single intracerebroventricular administration of 3 microg of IMC-A1 via the 3rd ventricle to mice caused an approximately 36% reduction in food intake and an approximately 6% weight loss within the ensuing 24 h. Our data suggest that FGF signaling through FGFR1c may play a physiological role in hypothalamic feeding circuit and that blocking it leads to hypophagia and weight loss.
Collapse
Affiliation(s)
- Haijun D Sun
- ImClone Systems Inc., 180 Varick St., New York, NY 10014, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Weickert CS, Kittell DA, Saunders RC, Herman MM, Horlick RA, Kleinman JE, Hyde TM. Basic fibroblast growth factor and fibroblast growth factor receptor-1 in the human hippocampal formation. Neuroscience 2005; 131:219-33. [PMID: 15680705 DOI: 10.1016/j.neuroscience.2004.09.070] [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] [Accepted: 09/01/2004] [Indexed: 11/23/2022]
Abstract
Basic fibroblast growth factor (bFGF) is an important mitogen and neurotrophic factor that binds and signals through the high-affinity receptor, fibroblast growth factor receptor 1 (FGFR1). However, only a limited amount of information is available concerning the molecular forms and anatomical distribution of fibroblast growth factors (FGFs) in the normal human brain. We found multiple bFGF and FGFR1 mRNA transcripts which vary in expression pattern across human brain regions. Using in situ hybridization and immunohistochemistry, we localized bFGF and FGFR1 mRNA and protein to cells in the normal adult human hippocampus and caudal entorhinal cortex (ERC). The majority of pyramidal neurons contained FGFR1 mRNA and protein in the mesial temporal lobe, with neurons in the CA2/CA3 region demonstrating the highest levels of FGFR1 mRNA. In contrast to FGFR1, bFGF mRNA expression was detected at very low levels in a small fraction of the neurons in the human hippocampus and caudal ERC. While bFGF mRNA may be expressed at low levels in neurons, bFGF-immunopositive cells with astrocytic features were detected throughout the mesial temporal lobe in rats, monkeys and humans. bFGF immunoreactive processes are found traversing the dentate gyrus, and bFGF immunoreactive cells are found in the neurogenic subgranular zone in all three mammalian species studied. The anatomical distribution of these two FGF family members suggests that bFGF is endogenously positioned to be involved in ongoing neurogenesis in the adult hippocampus, and that FGF trophic signaling to differentiated neurons could involve the release of astrocytic bFGF acting on neuronal FGFR1 in the normal adult human hippocampus.
Collapse
Affiliation(s)
- C S Weickert
- Neuropathology Section, Clinical Brain Disorders Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892-1385, USA.
| | | | | | | | | | | | | |
Collapse
|
14
|
Ohmachi S, Mikami T, Konishi M, Miyake A, Itoh N. Preferential neurotrophic activity of fibroblast growth factor-20 for dopaminergic neurons through fibroblast growth factor receptor-1c. J Neurosci Res 2003; 72:436-43. [PMID: 12704805 DOI: 10.1002/jnr.10592] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Degeneration of dopaminergic neurons of the substantia nigra causes Parkinson's disease. Therefore, neurotrophic factors for dopaminergic neurons are of substantial clinical interest. Fibroblast growth factor (FGF)-20 preferentially expressed in the substantia nigra pars compacta (SNPC) of the rat brain significantly enhanced the survival of midbrain dopaminergic neurons. Here we examined the mechanism of action of FGF-20 on dopaminergic neurons. FGF-20 slightly enhanced the survival of total neurons of the midbrain, indicating that it preferentially enhanced the survival of dopaminergic neurons. FGF receptor (FGFR)-1c was found to be expressed abundantly in dopaminergic neurons in the SNPC but at much lower levels in neurons of other midbrain regions by in situ hybridization. FGF-20 was also found to bind FGFR-1c with high affinity with the BIAcore system. Furthermore, FGF-20 activated the mitogen-activated protein kinase (MAPK) pathway, which is the major intracellular signaling pathway of FGFs. Both the FGFR-1 inhibitor SU5402 and the MAPK pathway inhibitor PD98059 also significantly inhibited the activation of the MAPK pathway by FGF-20 and the neurotrophic activity of FGF-20. The present findings indicate that the activation of the MAPK pathway by FGF-20 signaling through FGFR-1c plays important roles in the survival of dopaminergic neurons in the SNPC.
Collapse
Affiliation(s)
- Shigeki Ohmachi
- Department of Genetic Biochemistry, Kyoto University Graduate School of Pharmaceutical Sciences, Sakyo, Kyoto, Japan
| | | | | | | | | |
Collapse
|
15
|
Iwami M, Tooyama I, Kinoshita A, Matsuo A, Oomura Y, Sasaki K, Kimura H. Demonstration of Fibroblast Growth Factor Receptor-1 in Rat Adrenal Gland as Revealed by Reverse Transcription-polymerase Chain Reaction and Immunohistochemistry. Acta Histochem Cytochem 2003. [DOI: 10.1267/ahc.36.353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Mika Iwami
- Molecular Neuroscience Research Center, Shiga University of Medical Science
- Department of Pediatric Medicine, Shiga University of Medical Science
| | - Ikuo Tooyama
- Molecular Neuroscience Research Center, Shiga University of Medical Science
| | - Ayae Kinoshita
- Molecular Neuroscience Research Center, Shiga University of Medical Science
- Harvard Medical School, Alzheimer Research Unit, Massachusetts General Hospital
| | - Akinori Matsuo
- Molecular Neuroscience Research Center, Shiga University of Medical Science
| | - Yutaka Oomura
- Institute of Bio-Active Science, Nippon Zoki Pharmaceutical Co. Ltd
| | - Kazuo Sasaki
- Division of Bio-Information Engineering, Faculty of Engineering, Toyama University
| | - Hiroshi Kimura
- Molecular Neuroscience Research Center, Shiga University of Medical Science
| |
Collapse
|
16
|
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: 2.4] [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
|
17
|
Draghia-Akli R, Khan AS, Cummings KK, Parghi D, Carpenter RH, Brown PA. Electrical enhancement of formulated plasmid delivery in animals. Technol Cancer Res Treat 2002; 1:365-72. [PMID: 12625762 DOI: 10.1177/153303460200100507] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Electroporation has been shown to significantly increase plasmid transfer to the skeletal muscle, but this procedure is also implicated in muscle damage. We are reporting a highly efficient in vivo transfer of a plasmid formulated with poly-(L-glutamate) (PLG) into murine, canine and porcine muscle fibers using electric pulses of low field intensity. In mice and pigs, the use of secreted embryonic alkaline phosphatase (SEAP) as the indicator gene caused increased PLG expression by 2-3 fold compared to naked plasmid; while delivery of a PLG-plasmid formulation to dogs showed a 10-fold increase in serum SEAP levels compared to plasmid alone. Muscle lesions were reduced by the protective PLG. Thus, PLG may constitute a useful adjuvant for increased expression and reduced muscle trauma to plasmid DNA delivered by electroporation.
Collapse
Affiliation(s)
- Ruxandra Draghia-Akli
- Advisys, Inc., 2700 Research Forest Drive, Suite 180, The Woodlands, Texas 77381, USA.
| | | | | | | | | | | |
Collapse
|
18
|
Martens DJ, Seaberg RM, van der Kooy D. In vivo infusions of exogenous growth factors into the fourth ventricle of the adult mouse brain increase the proliferation of neural progenitors around the fourth ventricle and the central canal of the spinal cord. Eur J Neurosci 2002; 16:1045-57. [PMID: 12383233 DOI: 10.1046/j.1460-9568.2002.02181.x] [Citation(s) in RCA: 190] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Stem cells isolated from the fourth ventricle and spinal cord form neurospheres in vitro in response to basic fibroblast growth factor (FGF2)+heparin (H) or epidermal growth factor (EGF)+FGF2 together. To determine whether these growth factor conditions are sufficient to induce stem cells within the fourth ventricle and spinal cord to proliferate and expand their progeny in vivo, we infused EGF and FGF2, alone or together, with or without H, into the fourth ventricle for 6 days via osmotic minipumps. Animals were injected with bromodeoxyuridine (BrdU) on days 4, 5 and 6 of infusion in order to label cells proliferating in response to the growth factors. Infusions of EGF+FGF2+H into the fourth ventricle resulted in the largest proliferative effect, a 10.8-fold increase in the number of BrdU+ cells around the fourth ventricle, and a 33.5-fold increase in the number of BrdU+ cells around the central canal of the spinal cord, as compared to vehicle infused controls. The majority of the cells were nestin+ after 6 days of infusion. Seven weeks post-infusion, 22 and 30% of the number of BrdU+ cells induced to proliferate after 6 days of EGF+FGF2+H infusions were still detected around the fourth ventricle and central canal of the spinal cord, respectively. Analysis of the fates of the remaining cells showed that a small percentage of BrdU+ cells around the fourth ventricle and in the white matter of the spinal cord differentiated into astrocytes and oligodendrocytes. BrdU+ neurons were not found in the brainstem or in the grey matter of the cervical spinal cord 7 weeks post-infusion. These results show that endogenous stem cells and progenitors around the fourth ventricle and central canal of the spinal cord proliferate in response to exogenously applied growth factors, but unlike in the lateral ventricle where they generate some new neurons, they only produce new astrocytes and oligodendrocytes at 7 weeks post-infusion.
Collapse
Affiliation(s)
- David J Martens
- Department of Anatomy and Cell Biology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada.
| | | | | |
Collapse
|
19
|
Kravtsov AN, Oomura Y, Sudakov KV. Impulse activity of individual sensorimotor cortex neurons in rabbits after microiontophoretic administration of antibodies against acid fibroblast growth factor. PATHOPHYSIOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY FOR PATHOPHYSIOLOGY 2002; 8:255-258. [PMID: 12100970 DOI: 10.1016/s0928-4680(02)00013-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
We studied the sensitivity of sensorimotor cortex neurons in rabbits to microiontophoretic administration of antibodies against acid fibroblast growth factor (anti-aFGF). Spontaneous activity and reaction of neurons to electrical stimulation of 'feeding centers' in the lateral hypothalamic area (LHA) were recorded. We analyzed impulse activity of 52 neurons in the sensorimotor cortex. It was shown that 14 neurons (25%) reacted to microiontophoresis of anti-aFGF (excitation and inhibition in 9 and 5 neurons, respectively). Microiontophoretic administration of anti-aFGF did not change the reaction (excitation or inhibition) of 27 neurons to electrical stimulation of LHA. Initially, 14 neurons did not response to LHA stimulation. After microiontophoretic administration of anti-aFGF, 6 of 14 neurons displayed pronounced reactions to electrical stimulation of LHA (excitation and inhibition in 2 and 4 neurons, respectively). These data suggest that aFGF plays an important physiological role in feeding motivations.
Collapse
Affiliation(s)
- Alexander Nikolaevih Kravtsov
- P.K. Anokhin Institute of Normal Physiology, Russian Academy of Medical Sciences, 6/4, Mokhovaya street, 103009, Moscow, Russia
| | | | | |
Collapse
|
20
|
Goddard DR, Berry M, Kirvell SL, Butt AM. Fibroblast growth factor-2 induces astroglial and microglial reactivity in vivo. J Anat 2002; 200:57-67. [PMID: 11833655 PMCID: PMC1570884 DOI: 10.1046/j.0021-8782.2001.00002.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A role for fibroblast growth factor-2 (FGF-2) has been proposed in mediating the glial response to injury in the central nervous system (CNS). We have tested this possibility in vivo, by injecting FGF-2 into the cerebrospinal fluid (CSF) of the brain ventricles of young rats and analysing glial cells in the anterior medullary velum (AMV), which partly roofs the IVth ventricle. FGF-2 was administered at two different doses, low FGF-2 (500 ng mL(-1) CSF) and high FGF-2 (10 microg mL(-1) CSF), and saline vehicle was injected in controls. Injections were performed twice daily for three days, commencing at postnatal day (P) 6, and AMV were analysed at P9, using immunohistochemistry and Western blotting. Glial cells were unaffected by treatment with saline or low FGF-2, whereas high FGF-2 induced reactive changes in glial cell types: (1) there was increased GFAP expression in astrocytes, demonstrated by Western blot and immunohistochemistry, and astrocytes appeared hypertrophic, with increased process thickness and number; (2) the number of ED1 labelled microglia/macrophages was doubled, from 47 +/- 6 to 114 +/- 17 cells per field (0.75 mm2; values are mean +/- SEM), and microglia appeared activated, with a multipolar and granular appearance; (3) NG2 positive glial cells appeared more fibrous and there was increased density of processes, although there was no significant increase in their number; (4) oligodendrocyte somata were enlarged and there was a loss of myelin sheaths. The results show that at high CSF titres of FGF-2 induce glial reactivity in vivo and support a role for FGF-2 in the pathology of CNS injury and EAE.
Collapse
|
21
|
Hotta M, Kuriyama H, Arai K, Takano K, Shibasaki T. Fibroblast growth factor inhibits locomotor activity as well as feeding behavior of rats. Eur J Pharmacol 2001; 416:101-6. [PMID: 11282118 DOI: 10.1016/s0014-2999(01)00767-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The effects of acute and chronic intracerebroventricular (i.c.v.) administration of basic fibroblast growth factor (bFGF) on behavior were examined in free-feeding rats. An i.c.v. injection of bFGF induced behavioral changes, such as an increase in resting and decreases in grooming, moving, and food intake at a dose of 20 or 50 ng. These effects appeared at 4-5 h and lasted at least 11 h after the injection. These changes, as well as inhibition of body weight gain, were also found during a 6-day period of chronic i.c.v. infusion of bFGF at a dose of 20 ng/h. These results indicate that bFGF as both bolus i.c.v. injection and chronic i.c.v. infusion inhibits not only feeding behavior but also locomotor activity in rats. It is suggested that the inhibitory effect of bFGF on food intake may be in part ascribed to the suppression of behavior by bFGF.
Collapse
Affiliation(s)
- M Hotta
- Department of Medicine, Institute of Clinical Endocrinology, School of Medicine, Tokyo Women's Medical University, 8-1 Kawada-cho, Tokyo 162-8666, Shinjuku, Japan.
| | | | | | | | | |
Collapse
|
22
|
Suzuki S, Li AJ, Akaike T, Imamura T. Intracerebroventricular infusion of fibroblast growth factor-1 increases Fos immunoreactivity in periventricular astrocytes in rat hypothalamus. Neurosci Lett 2001; 300:29-32. [PMID: 11172932 DOI: 10.1016/s0304-3940(01)01535-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
When released into the third ventricle of the brain, fibroblast growth factor (FGF)-1 acts as a feeding suppressor. To identify the cells that respond to FGF-1 in this process, we investigated the effects of intracerebroventricular administration of FGF-1 on Fos protein expression in the rat hypothalamus. Two hours after infusion of FGF-1, significantly more Fos signals were observed in the periventricular zone of the third ventricle and its surroundings. Double immunohistochemical studies using antibodies against Fos, glial fibrillary acidic protein, and neuronal nuclei revealed that the FGF-1-induced Fos signals in the periventricular zone were in astrocytes, not in neurons. FGF-1-induced Fos expression was not found in neurons in such hypothalamic nuclei as the lateral hypothalamic area, paraventricular nucleus, ventromedial hypothalamic nucleus, dorsomedial hypothalamic nucleus, or arcuate nucleus. These results suggest a possible involvement of periventricular astrocytes in the early stages of FGF-1-induced feeding suppression.
Collapse
Affiliation(s)
- S Suzuki
- Biosignaling Department, National Institute of Bioscience and Human Technology, AIST, 1-1 Higashi, Ibaraki 305-8566, Tsukuba, Japan
| | | | | | | |
Collapse
|
23
|
Shiraishi T, Oomura Y, Sasaki K, Wayner MJ. Effects of leptin and orexin-A on food intake and feeding related hypothalamic neurons. Physiol Behav 2000; 71:251-61. [PMID: 11150556 DOI: 10.1016/s0031-9384(00)00341-3] [Citation(s) in RCA: 100] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The lateral hypothalamic area (LHA) and the ventromedial hypothalamic nucleus (VMH) have historically been implicated in ingestive behavior, energy balance and body mass regulation. The LHA is more closely associated with the initiation of eating; whereas the VMH mediates the cessation of eating. The parvocellular part of the paraventricular nucleus (pPVN) is also included in the suppressing mechanism. Recently, two hypothalamic peptides, orexin-A and orexin-B, localized in the posterior and lateral hypothalamic perifornical region were discovered in the rat brain and they increase food intake. Leptin, a protein encoded by an obesity gene, expressed in adipose tissue and released into the blood also affects food intake. Orexin and leptin receptors have been localized in the LHA, pPVN, and VMH. The purpose of this study was to measure food intake in the rat in response to leptin and orexin-A; and to determine their electrophysiological effects on feeding related hypothalamic neurons. Results clearly show that leptin suppresses food intake whereas orexin-A increases food intake. These differences are associated with leptin and orexin-A modulatory effects on LHA, pPVN, and VMH glucose responding neurons. In the LHA, leptin inhibits a larger proportion of both glucose-sensitive neurons (GSNs) and non-GSNs. In the pPVN, leptin increases more GSNs in comparison to non-GSNs. Whereas in the VMH, leptin increases the activity of glucoreceptor neurons (GRNs) in comparison to non-GRNs. Orexin-A had opposite effects: increases activity of GSNs more than the non-GSNs in the LHA and significantly suppresses GRNs in the VMH. In the pPVN, orexin-A had no observable effects on neurons that have a low density of orexin 2 receptors. Results are discussed in terms of hypothalamic neural circuits that are sensitive to endogenous food intake inducing and reducing substances.
Collapse
Affiliation(s)
- T Shiraishi
- Department of Physiology, Tokai University School of Medicine, Ishehara, 259-1193, Japan
| | | | | | | |
Collapse
|
24
|
Long-term rAAV-mediated gene transfer of GDNF in the rat Parkinson's model: intrastriatal but not intranigral transduction promotes functional regeneration in the lesioned nigrostriatal system. J Neurosci 2000. [PMID: 10844038 DOI: 10.1523/jneurosci.20-12-04686.2000] [Citation(s) in RCA: 296] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Previous studies have used recombinant adeno-associated viral (rAAV) vectors to deliver glial cell line-derived neurotrophic factor (GDNF) in the substantia nigra to protect the nigral dopamine (DA) neurons from 6-hydroxydopamine-induced damage. However, no regeneration or functional recovery was observed in these experiments. Here, we have used an rAAV-GDNF vector to express GDNF long-term (6 months) in either the nigral DA neurons themselves, in the striatal target cells, or in both of these structures. The results demonstrate that both nigral and striatal transduction provide significant protection of nigral DA neurons against the toxin-induced degeneration. However, only the rats receiving rAAV-GDNF in the striatum displayed behavioral recovery, accompanied by significant reinnervation of the lesioned striatum, which developed gradually over the first 4-5 months after the lesion. GDNF transgene expression was maintained at high levels throughout this period. These results provide evidence that rAAV is a highly efficient vector system for long-term expression of therapeutic proteins in the nigrostriatal system.
Collapse
|
25
|
Gremo F, Presta M. Role of fibroblast growth factor-2 in human brain: a focus on development. Int J Dev Neurosci 2000; 18:271-9. [PMID: 10715581 DOI: 10.1016/s0736-5748(99)00095-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Trophic factors have gained a great degree of attention as regulators of neural cells proliferation and differentiation as well as of brain maturation. Very little is known, however, about their effects on human immature nervous system. In this paper, data on expression of fibroblast-growth factor-2 and its receptors are reviewed and discussed in the light of its possible role in human brain development.
Collapse
Affiliation(s)
- F Gremo
- Department of Cytomorphology, School of Medicine, Cagliari, Italy.
| | | |
Collapse
|
26
|
Matsuo A, Nakamura S, Akiguchi I. Immunohistochemical localization of glial cell line-derived neurotrophic factor family receptor alpha-1 in the rat brain: confirmation of expression in various neuronal systems. Brain Res 2000; 859:57-71. [PMID: 10720615 DOI: 10.1016/s0006-8993(99)02442-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The localization of glial cell line-derived neurotrophic factor (GDNF) family receptor alpha-1 (GFRalpha-1) was investigated in rat brain by immunohistochemistry using a polyclonal antibody against a specific sequence of the rat protein. For raising the antisera in rabbits, we synthesized the oligopeptide SDVFQQVEHISKGN that corresponds to residues 139 to 152 of rat GFRalpha-1. On immunospot assay, 0.5 microg/ml of an affinity-purified antibody was capable of detecting 7.8 pmol of the rat GFRalpha-1 oligopeptides. When rat brain homogenates were examined by Western blots, the antibody revealed two main bands with molecular weights of approximately 47 kDa and 53 kDa, corresponding to the known sizes of GFRalpha-1. Immunohistochemistry in rat brain demonstrated that GFRalpha-1-like immunoreactivity was present in neurons but not in glial cells. The localization of GFRalpha-1-like immunoreactivity was largely consistent with that of the corresponding GFRalpha-1 mRNA. Positive neurons were distributed widely in various brain regions, but were particularly abundant in such regions as the olfactory bulb, diagonal band, substantia innominata, zona incerta, substantia nigra, cerebellar cortex, nuclei of the cranial nerves including auditory system and spinal motoneurons. The present study showed that GFRalpha-1 in the normal central nervous system is expressed preferentially in certain multiple neuronal systems that include cholinergic system as well as dopaminergic system and motor neurons. As GFRalpha-1 protein was found in numerous brain structures, GDNF family ligands may have therapeutic application not only in degenerative diseases affecting in specific nervous systems, such as Parkinson's disease, amyotrophic lateral sclerosis and multiple system atrophy, but in diffusely damaging diseases like cerebrovascular diseases.
Collapse
Affiliation(s)
- A Matsuo
- Department of Neurology, Kyoto University, 54 Shougoinkawara-cho, Sakyo-ku, Japan.
| | | | | |
Collapse
|
27
|
Tooyama I, Kimura H. A protein encoded by an alternative splice variant of choline acetyltransferase mRNA is localized preferentially in peripheral nerve cells and fibers. J Chem Neuroanat 2000; 17:217-26. [PMID: 10697248 DOI: 10.1016/s0891-0618(99)00043-5] [Citation(s) in RCA: 87] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Central cholinergic systems have been visualized by immunohistochemistry using antibodies to choline acetyltransferase (ChAT). Peripheral cholinergic cells and fibers, however, have been hardly detectable with most of these antibodies. This phenomenon suggests that a different form of ChAT may exist in peripheral tissues. Here we report two types of mRNA for ChAT expressed by alternative splicing in rat pterygopalatine ganglion. One is exactly identical with ChAT mRNA reported in the central nervous system (ChAT of a common type; cChAT). The other lacks exons 6, 7, 8 and 9, which was detected only in the pterygopalatine ganglion (ChAT of a peripheral type; pChAT). The peculiarity of pChAT in chemical structure, possessing a splice joint of the exons 5 and 10, led us to produce rabbit antisera against a recombinant peptide of 41 amino acids which spans over the splice joint. On Western blots using a successfully obtained antiserum, an intense band of about 50 kDa, corresponding to the expected molecular weight of pChAT, was detected in the pterygopalatine ganglion but not in the brain. Immunohistochemistry using the antiserum failed to reveal positive staining of known brain cholinergic structures, while it permitted us to observe peripheral, probably cholinergic, nerve cells and fibers including those in the pterygopalatine ganglion and enteric nervous system.
Collapse
Affiliation(s)
- I Tooyama
- Neuroanatomny Unit, Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu, Japan.
| | | |
Collapse
|
28
|
Mandel RJ, Rendahl KG, Snyder RO, Leff SE. Progress in direct striatal delivery of L-dopa via gene therapy for treatment of Parkinson's disease using recombinant adeno-associated viral vectors. Exp Neurol 1999; 159:47-64. [PMID: 10486174 DOI: 10.1006/exnr.1999.7159] [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: 11/22/2022]
Abstract
Viral vectors have recently been used successfully to transfer genes and express different proteins in the brain. This review discusses the requirements to consider human clinical trials in which recombinant adeno-associated virus vectors are used to transfer the genes necessary to produce l-dihydroxyphenylalanine (l-dopa) directly into the striatum of Parkinson's patients. Preclinical data that apply to the criteria defined as prerequisite for clinical trials are discussed. Thus, in animal models using recombinant adeno-associated virus vectors it has been demonstrated that l-dopa can be synthesized in the striatum after in vivo transduction. In addition, these l-dopa levels are sufficient to affect behavior in a dopamine-deficient animal model, the expression is extremely long-lasting, and the ability to transcriptionally regulate tyrosine hydroxylase has been demonstrated but not fully characterized. However, while immune responses to recombinant adeno-associated virus infection in the periphery have been studied, direct assessment of the potential immune response in the brain has not been sufficiently defined. Therefore, the rationale for delivering l-dopa directly to the striatum to treat Parkinson's disease is sound and the preclinical data are promising but all the issues surrounding this strategy are not resolved.
Collapse
Affiliation(s)
- R J Mandel
- Department of Neuroscience, University of Florida Brain Institute, University of Florida School of Medicine, P.O. Box 10024, Gainesville, FL, 32610-0244, USA.
| | | | | | | |
Collapse
|
29
|
Adult mammalian forebrain ependymal and subependymal cells demonstrate proliferative potential, but only subependymal cells have neural stem cell characteristics. J Neurosci 1999. [PMID: 10341247 DOI: 10.1523/jneurosci.19-11-04462.1999] [Citation(s) in RCA: 366] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The adult derivatives of the embryonic forebrain germinal zones consist of two morphologically distinct cell layers surrounding the lateral ventricles: the ependyma and the subependyma. Cell cycle analyses have revealed that at least two proliferating populations exist in this region, one that is constitutively proliferating and one that is relatively quiescent and thought to include the endogenous adult neural stem cells. Earlier studies demonstrated that specific dissection of the region surrounding the lateral ventricles was necessary for the in vitro isolation of multipotent, self-renewing neural stem cells. However, in these studies, the ependymal layer was not physically separated from the subependymal layer to identify the specific adult laminar localization of the neural stem cells around the lateral ventricles. To determine which cellular compartment in the adult forebrain contained the neural stem cells, we isolated and cultured the ependyma separately from the subependyma and tested for the presence of neural stem cells using the in vitro neurosphere assay. We demonstrate that the ependymal cells can proliferate in vitro to form sphere-like structures. However, the ependymal cells generating spheres do not have the ability to self-renew (proliferate to form secondary spheres after dissociation) nor to produce neurons, but rather only seem to generate glial fibrillary acidic protein-positive ependymal cells when plated under differentiation conditions in culture. On the other hand, a subpopulation of subependymal cells do possess the self-renewing and multipotential characteristics of neural stem cells. Therefore, the adult forebrain neural stem cell resides within the subependymal compartment.
Collapse
|
30
|
Li AJ, Oomura Y, Sasaki K, Suzuki K, Hori T. Protective effect of acidic fibroblast growth factor against ischemia-induced learning and memory deficits in two tasks in gerbils. Physiol Behav 1999; 66:577-83. [PMID: 10386900 DOI: 10.1016/s0031-9384(98)00330-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The influence of transient forebrain ischemia on behavioral performance, and the effect of intracerebroventricular (i.c.v.) injection of acidic fibroblast growth factor (aFGF) on such ischemia-induced deficits were examined in Mongolian gerbils by assessing learning and memory in two tasks: passive avoidance and Morris water maze. A 5-min period of forebrain ischemia led to learning and memory deficits in both tasks, and also to neuronal death in the hippocampal CA1 region. Continuous i.c.v. infusion of aFGF bilaterally into the lateral ventricules by osmotic minipumps over 2 days before, and 5 days after the ischemia (a total of 3.6 microg/gerbil) largely prevented both the ischemia-induced behavioral deficits and the neuronal death in the hippocampus. These observations suggest that the hippocampus is a critical site for the performance of the two tasks, and that aFGF has a protective effect against such ischemia-induced learning and memory deficits in gerbils.
Collapse
Affiliation(s)
- A J Li
- Department of Physiology, Faculty of Medicine, Kyushu University, Fukuoka, Japan.
| | | | | | | | | |
Collapse
|
31
|
Li AJ, Ozawa K, Tsuboyama H, Imamura T. Distribution of fibroblast growth factor-5 in rat hypothalamus, and its possible role as a regulator of feeding behaviour. Eur J Neurosci 1999; 11:1362-8. [PMID: 10103131 DOI: 10.1046/j.1460-9568.1999.00546.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We previously reported that a transcript of fibroblast growth factor-5 (FGF-5) was more abundant in the brain of postnatal and adult mice than in the embryonic brain. This suggested that FGF-5 plays some role in the mature brain. Here, we have investigated the spatiotemporal expression and function of FGF-5 in the adult rat hypothalamus with the emphasis on feeding behaviour. In situ hybridization experiments demonstrated that, in both adequately fed and fasted (20 h) rats, FGF-5 transcripts were present within several nuclei in the hypothalamus (viz. the magnocellular part of the paraventricular nucleus, supraoptic nucleus, arcuate nucleus, median eminence, and ventromedial hypothalamic nucleus), but not in the lateral hypothalamic area. Quantitative detection of FGF-5 mRNA in the hypothalamus (especially in the paraventricular nucleus) indicated that food deprivation (20 h) reduced the expression of this gene to almost one-half of that seen in the control (fed) rats. The expression recovered to the control level after 1 h re-feeding, and this recovery persisted for several hours. Furthermore, FGF-5, when infused into the third ventricle, consistently reduced food intake, water intake and body weight gain, all in a dose-dependent manner. These results suggest that FGF-5 in the hypothalamus acts as a physiological regulator of feeding behaviour, and that its decreased expression during food deprivation may be important in stimulating appetite.
Collapse
Affiliation(s)
- A J Li
- Biosignaling Department, National Institute of Bioscience and Human- Technology, Tsukuba, Ibaraki, Japan
| | | | | | | |
Collapse
|
32
|
Li AJ, Tsuboyama H, Komi A, Ikekita M, Imamura T. Strong suppression of feeding by a peptide containing both the nuclear localization sequence of fibroblast growth factor-1 and a cell membrane-permeable sequence. Neurosci Lett 1998; 255:41-4. [PMID: 9839722 DOI: 10.1016/s0304-3940(98)00701-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Earlier studies have shown that fibroblast growth factor (FGF)-1 in the brain regulates feeding behavior. In the present study, food intake in rats was strongly suppressed by an infusion into the lateral cerebroventricle of a synthetic peptide (26 amino acids) which contains both the N-terminal nuclear localization sequence (NLS) of FGF-1 and a recently identified membrane-permeable sequence. When the NLS motif in the peptide was destroyed by mutations of two lysine residues, the mutant peptide failed to affect eating. The results suggest that the NLS of FGF-1 plays an important role in FGF-1-induced feeding suppression and they introduce a novel compound for feeding regulation.
Collapse
Affiliation(s)
- A J Li
- Biosignaling Department, National Institute of Bioscience and Human Technology, Tsukuba, Japan
| | | | | | | | | |
Collapse
|
33
|
Takami K, Matsuo A, Terai K, Walker DG, McGeer EG, McGeer PL. Fibroblast growth factor receptor-1 expression in the cortex and hippocampus in Alzheimer's disease. Brain Res 1998; 802:89-97. [PMID: 9748519 DOI: 10.1016/s0006-8993(98)00552-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Localization of fibroblast growth receptor (FGFR)-1 immunoreactivity was investigated immunochemically in postmortem brain tissue of Alzheimer's disease (AD) and age-matched control cases using a rabbit polyclonal antibody and a mouse monoclonal antibody specific for FGFR-1. In control cases, FGFR-1 immunoreactivity was identified in astrocytes in white matter and in hippocampal pyramidal neurons. In AD cases, the immunoreactivity in reactive astrocytes surrounding senile plaques was increased. The pattern of FGFR-1 immunoreactivity was confirmed in selected cases by in situ hybridization for FGFR-1 mRNA. Immunoreactivity using a monoclonal antibody demonstrated a similar distribution pattern. The localization of FGFR-1 is consistent with previous reports on the involvement of FGF-1 and FGF-2 in AD.
Collapse
Affiliation(s)
- K Takami
- Kinsmen Laboratory of Neurological Research, University of British Columbia, Vancouver, Canada
| | | | | | | | | | | |
Collapse
|
34
|
Li AJ, Oomura Y, Sasaki K, Suzuki K, Tooyama I, Hanai K, Kimura H, Hori T. A single pre-training glucose injection induces memory facilitation in rodents performing various tasks: contribution of acidic fibroblast growth factor. Neuroscience 1998; 85:785-94. [PMID: 9639272 DOI: 10.1016/s0306-4522(97)00630-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Effects of a pre-training intraperitoneal glucose injection on learning and memory were tested using two tasks: passive avoidance and Morris water maze. In the former task, mice that had received glucose 2 h prior (but not 1, 3, or 5 h prior) to a trial that combined acquisition with passive avoidance of foot shock showed a significantly increased retention latency when tested 24 h later. Thus, this effect was time-dependent, and it was also found to be dose-dependent by further experiment. In contrast, 2-deoxy-D-glucose and fructose had no such effect. In the Morris water maze task, glucose injection 2 or 3 h before a block of trials enhanced the spatial memory performance of mice. These glucose-induced memory-facilitation effects were abolished by an intracerebroventricular injection of anti-acidic fibroblast growth factor antibody 30 min before the glucose injection, suggesting a critical role for endogenous acidic fibroblast growth factor in this facilitatory effect. Furthermore, continuous intracerebroventricular infusion of acidic fibroblast growth factor in rats significantly increased retention latency (when tested repeatedly on successive days using a passive avoidance task). Our earlier studies demonstrated that brain acidic fibroblast growth factor is produced in the ependymal cells of the cerebroventricular system, and is released into the cerebrospinal fluid following either a meal or a (intraperitoneal or intracerebroventricular) glucose injection. This released acidic fibroblast growth factor also diffuses into the brain parenchyma, and is taken up by neurons in the hippocampus, hypothalamus, and elsewhere in the brain some 2 h after the meal or glucose injection. These and the present findings indicate (i) that pre-training glucose injection improves memory performance, and (ii) that acidic fibroblast growth factor, especially by its action within the hippocampus, is involved in this enhancement process.
Collapse
Affiliation(s)
- A J Li
- Department of Physiology, Faculty of Medicine, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Walker DG, Terai K, Matsuo A, Beach TG, McGeer EG, McGeer PL. Immunohistochemical analyses of fibroblast growth factor receptor-1 in the human substantia nigra. Comparison between normal and Parkinson's disease cases. Brain Res 1998; 794:181-7. [PMID: 9622624 DOI: 10.1016/s0006-8993(98)00132-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The use of neurotrophic growth factors as a means of preventing loss of the dopaminergic (DA) neurons in the substantia nigra (SN) is becoming an accepted treatment strategy for Parkinson's disease (PD). In earlier studies, we showed that there was a selective loss of basic fibroblast growth factor (bFGF) immunoreactivity in DA neurons of the SN in PD suggesting that a deficiency of bFGF might contribute to cell death. As a preliminary step to assessing the potential for using bFGF or its analogs as therapeutic agents, the expression of fibroblast growth factor receptor-1 (FGFR-1) in the SN of normal and PD cases was investigated immunohistochemically. FGFR-1 immunoreactivity could be detected in DA neurons of the SN in young and old neurologically normal cases with an apparent decline with age. FGFR-1 immunoreactivity was also detected in many of the residual SN neurons in most of the idiopathic PD cases. These results indicate that FGFR-1 immunoreactivity, and possibly FGF binding activity, is retained in DA neurons in PD.
Collapse
Affiliation(s)
- D G Walker
- Kinsmen Laboratory of Neurological Research, Department of Psychiatry and Neurodegenerative Disease Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | | | | | |
Collapse
|
36
|
Abstract
A survey of the literature shows that proliferation of ependyma occurs largely during the embryonic and early postnatal periods of development in most species. Differentiation of these cells proceeds along particular regional and temporal gradients as does the expression of various cytoskeletal (vimentin, cytokeratins, glial fibrillary acidic protein) and secretory proteins (S-100). Turnover declines significantly postnatally, and only low levels of residual activity persist into adulthood under normal conditions. Although the reported response of ependyma to injury is somewhat equivocal, only limited regenerative capacity appears to exist and to varying degrees in different regions of the neuraxis. Proliferation has been most often observed in response to spinal cord injury. Indeed, the ependyma plays a significant role in the initiation and maintenance of the regenerative processes in the spinal cord of inframammalian vertebrates. In the human, however, ependyma appears never to regenerate at any age nor re-express cytoskeletal proteins characteristic of immature cells. The functions of ependyma including tanycytes, a specialized form of ependymal cell that persists into adulthood within circumscribed regions of the nervous system, are still largely speculative. Fetal unlike mature ependyma is believed to be secretory and is believed to play a role in neurogenesis, neuronal differentiation/axonal guidance, transport, and support. In the adult brain, mature ependyma is not merely an inert lining but may regulate the transport of ions, small molecules, and water between the cerebrospinal fluid and neuropil and serve an important barrier function that protects neural tissue from potentially harmful substances by mechanisms that are still incompletely understood.
Collapse
Affiliation(s)
- J E Bruni
- Department of Human Anatomy and Cell Science, The University of Manitoba, Winnipeg, Canada
| |
Collapse
|
37
|
Takenaka H, Kishimoto S, Tooyama I, Kimura H, Yasuno H. Protein expression of fibroblast growth factor receptor-1 in keratinocytes during wound healing in rat skin. J Invest Dermatol 1997; 109:108-12. [PMID: 9204964 DOI: 10.1111/1523-1747.ep12276740] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Fibroblast growth factors have been shown to play important roles in wound healing. To define their sites of action, we examined the expression of fibroblast growth factor receptor-1 (FGFR-1) during burn wound healing in rat skin by immunohistochemistry and western blot analysis. In cryostat sections of intact skin, little or no staining was observed. After a burn, however, staining for FGFR-1 was found in newly forming epidermis. The suprabasal layer of such epidermis, composed mostly of regenerating keratinocytes, was stained intensely, whereas keratinocytes in newly forming hair follicles were devoid of staining. Staining gradually decreased week by week after wound closure and was hardly visible 10 weeks after the burn, when the thickness of the epidermis had returned to the normal level. Staining was also found in small blood vessels and capillaries of granulation tissues of the dermis. Western blot analysis using the same antiserum was performed in the newly forming epidermis 10 d after the burn. A single band was detected with an apparent molecular weight of 120 kDa, corresponding to the short membrane-bound form of rat FGFR-1. Our study indicates that FGFR-1 is expressed during wound healing, mainly in regenerating epidermis and to some extent in blood vessels of the dermis. Fibroblast growth factors may affect the proliferation and differentiation of epidermal keratinocytes as well as angiogenesis in the dermis via the FGFR-1 expressed during wound healing.
Collapse
Affiliation(s)
- H Takenaka
- Institute of Molecular Neurology, Shiga University of Medical Science, Otsu, Japan
| | | | | | | | | |
Collapse
|
38
|
Inglis WL, Semba K. Discriminable excitotoxic effects of ibotenic acid, AMPA, NMDA and quinolinic acid in the rat laterodorsal tegmental nucleus. Brain Res 1997; 755:17-27. [PMID: 9163537 DOI: 10.1016/s0006-8993(97)00101-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Excitotoxins are valuable tools in neuroscience research as they can help us to discover the extent to which certain neurones are necessary for different types of behaviour. They have distinctive neurotoxic effects depending on where they are infused, and this study was conducted to delineate the neurotoxic profiles of excitotoxins in the laterodorsal tegmental nucleus (LDTg). Two 0.1 microl infusions of 0.1 M ibotenate, 0.1 M quinolinate, 0.04-0.1 M NMDA, or 0.05-0.015 M AMPA, were made unilaterally into the LDTg under either pentobarbitone or Avertin anaesthesia. The injection needle was oriented at an angle of 24 degrees from vertical in the mediolateral plane. After 23-27 days, sections through the mesopontine tegmentum were processed using standard histological procedures for NADPH-diaphorase histochemistry, tyrosine hydroxylase or 5-hydroxytryptamine immunohistochemistry, and Cresyl violet. Lesions were assessed in terms of the size of the damaged area (identified by reactive gliosis), the extent of cholinergic cell loss in the mesopontine tegmentum (by counting NADPH-diaphorase-positive neurones), and neuronal loss induced in the locus coeruleus and dorsal raphe nucleus. Ibotenate induced compact lesions in the LDTg (more than 80% cholinergic loss) and did little damage to the locus coeruleus and dorsal raphe nucleus. Quinolinate and low doses of AMPA and NMDA made very small lesions with less than 35% cholinergic loss, while at higher doses, AMPA and NMDA induced large areas of reactive gliosis but killed only a proportion of the cholinergic neurones. AMPA appeared to have a particular affinity for noradrenergic neurones in the locus coeruleus, with the 0.015 M dose injected into the LDTg typically destroying the majority of these neurones. The results are discussed in the context of what is known about the mechanisms of excitotoxins and the glutamate receptor profile of mesopontine neurones.
Collapse
Affiliation(s)
- W L Inglis
- Department of Anatomy and Neurobiology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | | |
Collapse
|
39
|
Abstract
If the eye lens of the adult newt, Notophthalmus viridescens, is removed, a new lens will regenerate and only from the dorsal, not the ventral, iris. The source, pigmented epithelial cells, would normally no longer divide, but upon lentectomy they do re-enter the cell cycle and form lens. The cause for this capability is unknown, but the mitogenic Fibroblast Growth Factors and their receptors may be involved. We have demonstrated that FGF receptors are present and operative in lens regeneration, since receptor-directed mitotoxins inhibit regeneration; heterogeneity and differential density in FGF-binding and receptor localization in iris sectors is also present. We propose that the spatial distribution of FGF receptors, especially the amphibian homolog of FGFR-3, is important in initiation of regeneration of eye lens.
Collapse
Affiliation(s)
- D S McDevitt
- Department of Animal Biology, University of Pennsylvania School of Veterinary Medicine, Philadelphia 19104, USA
| | | | | | | |
Collapse
|
40
|
Neurogenesis in the Adult Brain: Lessons Learned from the Studies of Progenitor Cells from the Embryonic and Adult Central Nervous Systems. ACTA ACUST UNITED AC 1997. [DOI: 10.1007/978-3-642-80308-6_8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
|
41
|
Blanquet PR, Jonet L. Signal-regulated proteins and fibroblast growth factor receptors: comparative immunolocalization in rat retina. Neurosci Lett 1996; 214:135-8. [PMID: 8878102 DOI: 10.1016/0304-3940(96)12921-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
We compared the immunolocalization of fibroblast growth factor (FGF)-R1 and FGF-R2 with that of several intracellular signalling proteins in rat neural retina. Only the serine/threonine extracellular signal-related kinases (ERK) and lipocortin/annexin 6, a major calcium-binding protein, appeared to be co-localized with FGF-R1 and FGF-R2 in all subfields of the neural retina. In particular, ERK appeared to be present in perikarya of ganglion cells and synaptic layers as did these receptors. Possible implications of these results with regard to neuroprotective role of FGF are discussed.
Collapse
Affiliation(s)
- P R Blanquet
- INSERM U450, Développment, Vieillissement et Pathologie de la rétine, Paris, France
| | | |
Collapse
|
42
|
Sasaki K, Oomura Y, Li AJ, Hanai K, Tooyama I, Kimura H, Yanaihara N, Hori T. Actions of acidic fibroblast growth factor fragments on food intake in rats. OBESITY RESEARCH 1995; 3 Suppl 5:697S-706S. [PMID: 8653551 DOI: 10.1002/j.1550-8528.1995.tb00488.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Acidic fibroblast growth factor (aFGF) has suppressive effects on food intake. In the present study, the effect of aFGF fragments on food intake were investigated in rats. Infusion of a carboxyl-terminal fragment of aFGF, aFGF-(114-140), did not affect food intake, whereas an amino-terminal fragment of aFGF, aFGF-(1-15), was significantly inhibitory. Other amino-terminal fragments, aFGF-(1-20), aFGF-(1-29) and aFGF-(9-29), did not affect food intake. However, [Ala16]aFGF-(1-29) and [Ser16]aFGF-(1-29) in which the cysteine residue at position 16 was replaced with alanine and serine, respectively, had significant suppressive effects on food intake. Infusion of a functional antagonist for FGF receptor, anti-FGFR-1 antibody, into the lateral hypothalamus (LHA) significantly increased food intake. The results suggest that: the amino-terminal portion of aFGF is active in food intake suppression; the replacement of cysteine residue by alanine or serine is important in some amino-terminal aFGF fragments; and the LHA is involved in feeding suppression actions by aFGF and some fragments.
Collapse
Affiliation(s)
- K Sasaki
- Division of Bio-Information Engineering, Faculty of Engineering, Toyama University, Japan
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Gonzalez AM, Berry M, Maher PA, Logan A, Baird A. A comprehensive analysis of the distribution of FGF-2 and FGFR1 in the rat brain. Brain Res 1995; 701:201-26. [PMID: 8925285 DOI: 10.1016/0006-8993(95)01002-x] [Citation(s) in RCA: 212] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
We have examined the cellular distribution of both FGF-2 and FGFR1 immunoreactivity and their mRNAs throughout the normal adult rat brain in order to reconcile numerous disparate findings in the published literature. The results confirm a widespread distribution of FGF-2 and FGFR1 in the rat brain, and different regions express distinct patterns of FGF-2 and FGFR1 mRNA and protein: neuronal and non-neuronal cells show different subcellular distributions that vary according to the area where they are located. The intensity of the staining and hybridization also varies according to the loci examined and the cell type involved. Astrocytes contain the highest levels of FGF-2 and FGFR1 mRNAs, and characteristically, possess high levels of immunoreactive FGF-2 within the nucleus. Amongst non-neuronal cells, oligodendrocytes do not synthesize or contain significant levels of FGF-2 immunoreactivity however, they do express FGFR1 mRNA. In these cells, immunoreactive FGFR1 is mainly associated with the myelin sheaths of neuronal fibers. In ventricular systems, ependymal cells synthesize and contain immunoreactive FGFR1. In contrast, only cells lining the lateral wall of the IIIrd ventricle express FGF-2 mRNA. Subependymal cells contain high levels of both FGF-2 and FGFR1 immunoreactivity. Neurons express low levels of FGF-2 mRNA and immunoreactive FGF-2 is localized predominantly to the perikaryon. However, selected populations of neurons, such as CA2 field of the hippocampus, show high levels of FGF-2 mRNA, in which the nucleus is strongly immunopositive. Similarly, high levels of FGFR1 mRNA are localized to select populations of neurons (e.g. amygdala). FGFR1 immunoreactivity is mainly associated with myelinated fiber tracts (e.g. striatum), and some neurons show immunoreactivity in the perikaryon (e.g. hippocampus), the nucleus (e.g. mesencephalic trigeminal nucleus), or in axonal projections (e.g. hypothalamus). Remarkably, in many of the areas studied, FGF-2 and FGFR1 mRNA and/or their translated protein do not co-localize in neurons (e.g. neo-cortices) or even in the same regions of the brain (e.g. substantia nigra). In other instances, mRNAs for both FGF-2 and FGFR1 colocalize (e.g. supraoptic nucleus). The brain, in contrast to peripheral tissues, contains high levels of FGF-2 and actively expresses its gene under normal physiological conditions. The highly specific anatomical distribution of immunoreactive FGF-2 in neuronal and non-neuronal brain cells, supports the notion that it plays a multifunctional role in the CNS under normal physiology. By correlating the localization and the synthesis of FGF-2 and one of its high affinity receptors, FGFR1, in the CNS, it should be possible to obtain a better understanding of the roles of FGF-2 in normal and pathological conditions.
Collapse
|
44
|
Williams EJ, Mittal B, Walsh FS, Doherty P. FGF inhibits neurite outgrowth over monolayers of astrocytes and fibroblasts expressing transfected cell adhesion molecules. J Cell Sci 1995; 108 ( Pt 11):3523-30. [PMID: 8586663 DOI: 10.1242/jcs.108.11.3523] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We have cultured cerebellar neurons on monolayers of cortical astrocytes in control medium or medium containing recombinant basic fibroblast growth factor (FGF). FGF was found to inhibit neurite outgrowth, with a significant effect seen at 0.5 ng/ml and a maximal effect at 10 ng/ml. FGF increased the production of arachidonic acid (AA) in cerebellar neurons, and when added directly to cultures or generated endogenously via activation of phospholipase A2 using melittin, this second messenger could mimic the inhibitory effect of FGF. FGF and AA could also specifically inhibit neurite outgrowth stimulated by three cell adhesion molecules (NCAM, N-cadherin and L1) expressed in transfected fibroblasts, or in the case of L1 bound to a tissue culture substratum. These data demonstrate that, in certain cellular contexts, FGF can act as an inhibitory cue for axonal growth and that arachidonic acid is the second messenger responsible for this activity. We discuss the possibility that arachidonic acid inhibits neurite outgrowth by desensitising the second messenger pathway underlying neuronal responsiveness to cell adhesion molecules.
Collapse
Affiliation(s)
- E J Williams
- Department of Experimental Pathology, UMDS, Guy's Hospital, London, UK
| | | | | | | |
Collapse
|
45
|
McMahon SB, Priestley JV. Peripheral neuropathies and neurotrophic factors: animal models and clinical perspectives. Curr Opin Neurobiol 1995; 5:616-24. [PMID: 8580713 DOI: 10.1016/0959-4388(95)80067-0] [Citation(s) in RCA: 80] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
A large body of data exists showing that a wide variety of neurotrophic factors can promote the survival or growth of different neuronal populations in vitro. More recently, several studies have been published on the survival-promoting effects of particular factors in animal models of peripheral neuropathies. Thus, the effect of axotomy on neuropeptide expression in dorsal root ganglion cells is partially reversed by nerve growth factor treatment, and the effect on choline acetyltransferase expression in motoneurones is partially reversed by glial-derived neurotrophic factor, neurotrophin-4/5 and brain-derived neurotrophic factor. Nerve growth factor also ameliorates some of the changes seen in sensory neurones in animal models of diabetic neuropathy and small fibre cytostatic drug neuropathy, whereas neurotrophin-3 has been found to reverse some changes in large sensory neurones associated with cisplatin neurotoxicity. The results of these studies provide grounds for optimism in the clinical uses of such factors, and, indeed, several clinical studies are now under way.
Collapse
Affiliation(s)
- S B McMahon
- Department of Physiology, UMDS, St Thomas' Hospital Medical School, London, UK.
| | | |
Collapse
|
46
|
Hamaguchi A, Tooyama I, Yoshiki T, Kimura H. Demonstration of fibroblast growth factor receptor-I in human prostate by polymerase chain reaction and immunohistochemistry. Prostate 1995; 27:141-7. [PMID: 7567692 DOI: 10.1002/pros.2990270304] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The expression and localization of fibroblast growth factor receptor-1 were investigated in human prostatic tissues with or without benign hyperplasia. Using a polymerase chain reaction method, we were able to demonstrate that prostatic tissues with benign hyperplasia expressed a significantly higher level of fibroblast growth factor receptor-1 mRNA than normal prostatic tissues (P < 0.01 by Anova). Western blot analysis using an antiserum against the receptor gave 2 bands with molecular weights of about 140 kDa and 80 kDa; these correspond to the expected sizes of the long and secreted forms of the fibroblast growth factor receptor-1, respectively. An immunohistochemical study using the same antiserum further demonstrated that the immunoreactive staining occurred mainly in the basal cells of the glandular epithelium and occasionally in the stromal cells. These results suggest that fibroblast growth factors may influence, at least in part, the proliferation of the epithelial cells seen in benign hyperplasia of human prostate.
Collapse
MESH Headings
- Adult
- Aged
- Animals
- Base Sequence
- Blotting, Western
- Brain/metabolism
- DNA Primers/analysis
- DNA Primers/chemistry
- DNA Primers/genetics
- DNA, Neoplasm/analysis
- DNA, Neoplasm/chemistry
- DNA, Neoplasm/genetics
- Epithelium/chemistry
- Epithelium/pathology
- Fibroblast Growth Factors/metabolism
- Humans
- Immunohistochemistry
- Male
- Middle Aged
- Molecular Sequence Data
- Polymerase Chain Reaction
- Prostate/chemistry
- Prostate/pathology
- Prostate/ultrastructure
- Prostatic Hyperplasia/metabolism
- Prostatic Hyperplasia/pathology
- RNA, Messenger/analysis
- RNA, Messenger/chemistry
- RNA, Messenger/genetics
- Rats
- Receptor Protein-Tyrosine Kinases
- Receptor, Fibroblast Growth Factor, Type 1
- Receptors, Fibroblast Growth Factor/analysis
- Receptors, Fibroblast Growth Factor/genetics
- Receptors, Fibroblast Growth Factor/metabolism
- Stromal Cells/chemistry
- Stromal Cells/pathology
Collapse
Affiliation(s)
- A Hamaguchi
- Department of Urology, Shiga University of Medical Science, Otsu, Japan
| | | | | | | |
Collapse
|
47
|
Lolova IS, Lolov SR. Age-related changes in basic fibroblast growth factor-immunoreactive cells of rat substantia nigra. Mech Ageing Dev 1995; 82:73-89. [PMID: 8538246 DOI: 10.1016/0047-6374(95)01599-u] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Immunohistochemistry and computer-assisted image analysis were used to examine the age-related changes in bFGF-immunoreactivity in rat substantia nigra (SN). Distribution pattern, number, size and staining intensity of bFGF-immunoreactive (bFGF-ir) cells in pars compacta and pars reticulata of 3-, 12- and 26-month-old rats were compared. The overall distribution of bFGF-immunoreactivity was similar in the three age groups, but changes in the morphological appearance of bFGF-ir somata and processes occurred in aging. The results demonstrated a significantly reduced number of bFGF-ir cells in pars compacta (by 56.87%) and pars reticulata (by 30.4%) in 26-month-old rats compared to 3-month-old rats. The reduction of the cell number did not occur smoothly and equally in the two parts of SN. The quantitative analysis clearly indicated a significant decrease in the size of bFGF-ir neurons in pars compacta (by 18.1%) and pars reticulata (by 14.15%) of 26-month-old rats compared to 3-month-old rats. Compared to 3-month-old rats, a 19.77% and 17.83% increase in the staining intensity was observed in the remaining bFGF-ir neurons of pars compacta and pars reticulata, respectively in 26-month-old rats. Since there was no correlation between the decreased size and increased staining, it is most probable that the intensification of the staining intensity of bFGF-ir neurons was a compensatory response to the cell death.
Collapse
Affiliation(s)
- I S Lolova
- Institute of Physiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | | |
Collapse
|
48
|
Oellig C, Pirvola U, Taylor L, Elde R, Hökfelt T, Pettersson RF. Acidic FGF and FGF receptors are specifically expressed in neurons of developing and adult rat dorsal root ganglia. Eur J Neurosci 1995; 7:863-74. [PMID: 7542125 DOI: 10.1111/j.1460-9568.1995.tb01073.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Employing complementary technical approaches, we have studied the expression of acidic fibroblast growth factor (aFGF) and FGF receptors in rat dorsal root ganglia. The results clearly showed that within spinal nerves aFGF and two high-affinity FGF receptors, FGFR-1 and FGFR-2, were prominently expressed in neurons, while expression in Schwann cells was undetectable. FGFR-3 and FGFR-4 were not expressed in dorsal root ganglia. Acidic FGF mRNA was detected in the majority of dorsal root ganglion neurons, including all size classes: FGFR-1 and FGFR-2 transcripts were only detected in subpopulations of mainly large and medium size neurons. In subcellular fractionation studies on dorsal root ganglion and spinal root tissue, aFGF was recovered in the soluble fraction and was thus not tightly associated with neuronal membranes. During development FGFR-1 and FGFR-2 mRNAs were found to be present at all stages examined (embryonic days 15-21 and postnatal days 1-120). Acidic FGF mRNA and protein were first detected at embryonic day 18, and their expression then increased progressively up to postnatal levels. In cultures of dorsal root ganglion neurons derived from day 15 embryos, aFGF expression was first detected 3 days after plating. The resulting neuron cultures continued to express aFGF in a Schwann cell-independent manner. In combination, these results indicate that aFGF expression in dorsal root ganglia is initiated and maintained in postmitotic neurons. Furthermore, the data suggest that the physiological function of aFGF in the peripheral nervous system is connected to processes specific to the mature sensory (and motor) system, such as the maintenance and survival of peripheral nerve neurons.
Collapse
Affiliation(s)
- C Oellig
- Ludwig Institute for Cancer Research, Stockholm Branch, Sweden
| | | | | | | | | | | |
Collapse
|
49
|
Abstract
This review summarizes the current scientific literature concerning the ependymal lining of the cerebral ventricles of the brain with an emphasis on selective barrier function and protective roles for the common ependymal cell. Topics covered include the development, morphology, protein and enzyme expression including reactive changes, and pathology. Some cells lining the neural tube are committed at an early stage to becoming ependymal cells. They serve a secretory function and perhaps act as a cellular/axonal guidance system, particularly during fetal development. In the mature mammalian brain ependymal cells possess the structural and enzymatic characteristics necessary for scavenging and detoxifying a wide variety of substances in the CSF, thus forming a metabolic barrier at the brain-CSF interface.
Collapse
Affiliation(s)
- M R Del Bigio
- Department of Pathology, Health Sciences Centre, Winnipeg, Canada
| |
Collapse
|
50
|
Sasaki K, Oomura Y, Figurov A, Morita N, Yanaihara N. An amino-terminal fragment peptide of acidic fibroblast growth factor modulates synaptic transmission in rat hippocampal slices. Brain Res Bull 1995; 38:185-91. [PMID: 7583346 DOI: 10.1016/0361-9230(95)00092-s] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Effects of acidic fibroblast growth factor (aFGF) fragments such as aminoterminal aFGF (1-15) and carboxyl-terminal aFGF (114-140) on synaptic transmission were investigated in rat hippocampal slices. Stimulation was applied to Schaffer collateral/commissural afferents, and evoked population spikes were recorded in the CA1 pyramidal cell layer. Continuous perfusion of slices with aFGF (1-15) slightly decreased the basal amplitude of population spikes and significantly increased the paired-pulse facilitation. When brief tetanic stimulation (7 pulses at 100 Hz) was applied 30 min after the perfusion of aFGF (1-15), aFGF (1-15)-treated slices enhanced the magnitude of short-term potentiation after the tetanus and facilitated a generation of long-term potentiation. These effects of aFGF (1-15) were dose-dependent. Perfusion of slices with aFGF (114-140) had no effect on the basal spike amplitude, paired-pulse facilitation, and short-term potentiation. Both aFGF (1-15) and aFGF (114-140) had no effect on the DNA synthesis-stimulating activity in BALB/c 3T3-L1 cells. The results suggest that aFGF (1-15), which is not involved in mitogenic activity, is implicated in a modulatory mechanism of synaptic plasticity.
Collapse
Affiliation(s)
- K Sasaki
- Division of Bio-Information Engineering, Faculty of Engineering, Toyama University, Japan
| | | | | | | | | |
Collapse
|