1
|
Medina-Arellano AE, Albert-Garay JS, Medina-Sánchez T, Fonseca KH, Ruiz-Cruz M, Ochoa-de la Paz L. Müller cells and retinal angiogenesis: critical regulators in health and disease. Front Cell Neurosci 2024; 18:1513686. [PMID: 39720707 PMCID: PMC11666533 DOI: 10.3389/fncel.2024.1513686] [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: 10/18/2024] [Accepted: 11/27/2024] [Indexed: 12/26/2024] Open
Abstract
Müller cells are the most abundant glial cells in the mammalian retina. Their morphology and metabolism enable them to be in close contact and interact biochemically and physically with almost all retinal cell types, including neurons, pericytes, endothelial cells, and other glial cells, influencing their physiology by releasing bioactive molecules. Studies indicate that Müller glial cells are the primary source of angiogenic growth factor secretion in the neuroretina. Because of this, over the past decade, it has been postulated that Müller glial cells play a significant role in maintaining retinal vascular homeostasis, with potential implications in vasoproliferative retinopathies. This review aims to summarize the current understanding of the mechanisms by which Müller glial cells influence retinal angiogenesis in health and disease, with a particular emphasis on three of the retinopathies with the most significant impact on visual health worldwide: diabetic retinopathy, retinopathy of prematurity, and age-related macular degeneration.
Collapse
Affiliation(s)
- Alan E. Medina-Arellano
- Laboratorio de Neurobiología Molecular y Celular de la Glía, Facultad de Medicina, Departamento de Bioquímica, UNAM, Mexico City, Mexico
- Unidad de Investigación APEC-UNAM, Asociación para Evitar la Ceguera en México I.A.P., Mexico City, Mexico
- Programa de Doctorado en Ciencias Biomédicas, UNAM, Mexico City, Mexico
| | - Jesús Silvestre Albert-Garay
- Laboratorio de Neurobiología Molecular y Celular de la Glía, Facultad de Medicina, Departamento de Bioquímica, UNAM, Mexico City, Mexico
- Unidad de Investigación APEC-UNAM, Asociación para Evitar la Ceguera en México I.A.P., Mexico City, Mexico
| | - Tania Medina-Sánchez
- Laboratorio de Neuroquímica, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría “Ramón de la Fuente Muñiz”, Mexico City, Mexico
| | - Karla Hernández Fonseca
- Laboratorio de Neuroquímica, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría “Ramón de la Fuente Muñiz”, Mexico City, Mexico
| | - Matilde Ruiz-Cruz
- Unidad de Investigación APEC-UNAM, Asociación para Evitar la Ceguera en México I.A.P., Mexico City, Mexico
| | - Lenin Ochoa-de la Paz
- Laboratorio de Neurobiología Molecular y Celular de la Glía, Facultad de Medicina, Departamento de Bioquímica, UNAM, Mexico City, Mexico
- Unidad de Investigación APEC-UNAM, Asociación para Evitar la Ceguera en México I.A.P., Mexico City, Mexico
| |
Collapse
|
2
|
Tang L, Xu GT, Zhang JF. Inflammation in diabetic retinopathy: possible roles in pathogenesis and potential implications for therapy. Neural Regen Res 2022; 18:976-982. [PMID: 36254977 PMCID: PMC9827774 DOI: 10.4103/1673-5374.355743] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Diabetic retinopathy, characterized as a microangiopathy and neurodegenerative disease, is the leading cause of visual impairment in diabetic patients. Many clinical features observed in diabetic retinopathy, such as capillary occlusion, acellular capillaries and retinal non-perfusion, aggregate retinal ischemia and represent relatively late events in diabetic retinopathy. In fact, retinal microvascular injury is an early event in diabetic retinopathy involving multiple biochemical alterations, and is manifested by changes to the retinal neurovascular unit and its cellular components. Currently, intravitreal anti-vascular endothelial growth factor therapy is the first-line treatment for diabetic macular edema, and benefits the patient by decreasing the edema and improving visual acuity. However, a significant proportion of patients respond poorly to anti-vascular endothelial growth factor treatments, indicating that factors other than vascular endothelial growth factor are involved in the pathogenesis of diabetic macular edema. Accumulating evidence confirms that low-grade inflammation plays a critical role in the pathogenesis and development of diabetic retinopathy as multiple inflammatory factors, such as interleukin-1β, monocyte chemotactic protein-1 and tumor necrosis factor -α, are increased in the vitreous and retina of diabetic retinopathy patients. These inflammatory factors, together with growth factors such as vascular endothelial growth factor, contribute to blood-retinal barrier breakdown, vascular damage and neuroinflammation, as well as pathological angiogenesis in diabetic retinopathy, complicated by diabetic macular edema and proliferative diabetic retinopathy. In addition, retinal cell types including microglia, Müller glia, astrocytes, retinal pigment epithelial cells, and others are activated, to secrete inflammatory mediators, aggravating cell apoptosis and subsequent vascular leakage. New therapies, targeting these inflammatory molecules or related signaling pathways, have the potential to inhibit retinal inflammation and prevent diabetic retinopathy progression. Here, we review the relevant literature to date, summarize the inflammatory mechanisms underlying the pathogenesis of diabetic retinopathy, and propose inflammation-based treatments for diabetic retinopathy and diabetic macular edema.
Collapse
Affiliation(s)
- Lei Tang
- Department of Ophthalmology of Tongji Hospital, Tongji Eye Institute, Department of Regenerative Medicine, and Department of Pharmacology, Tongji University School of Medicine, Shanghai, China
| | - Guo-Tong Xu
- Department of Ophthalmology of Tongji Hospital, Tongji Eye Institute, Department of Regenerative Medicine, and Department of Pharmacology, Tongji University School of Medicine, Shanghai, China,Correspondence to: Guo-Tong Xu, ; Jing-Fa Zhang, .
| | - Jing-Fa Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People’s Hospital), Shanghai Jiao Tong University, Shanghai, China,National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China,Correspondence to: Guo-Tong Xu, ; Jing-Fa Zhang, .
| |
Collapse
|
3
|
Araújo RS, Santos DF, Silva GA. The role of the retinal pigment epithelium and Müller cells secretome in neovascular retinal pathologies. Biochimie 2018; 155:104-108. [PMID: 29960032 DOI: 10.1016/j.biochi.2018.06.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 06/25/2018] [Indexed: 02/06/2023]
Abstract
Secreted trophic factors are key to maintain the structural and functional integrity of the retina, as they regulate cellular pathways responsible for survival, function, and response to injury. Nevertheless, these same factors can also be involved in retinal pathologies, as a consequence of the impairment of the secretory function of cells. The cells considered as major contributors to the retinal secretome are the retinal pigmented epithelium (RPE) and Müller cells. Their role in the pathophysiology of the most common neovascular pathologies in the retina - Age-related Macular Degeneration (AMD), Diabetic Retinopathy (DR), and Retinopathy of Prematurity (ROP) - is highlighted in this short review, together with current trophic factor-based therapies, which are mainly focused on controlling inflammation, cell survival, and angiogenesis.
Collapse
Affiliation(s)
- Rute S Araújo
- CEDOC - Chronic Diseases Center, NOVA Medical School, Universidade Nova de Lisboa, Campo Mártires da Pátria 130, 1169-056 Lisboa, Portugal; Bioengineering-Cell Therapies and Regenerative Medicine PhD Program, NOVA Medical School, Universidade Nova de Lisboa, Campo Mártires da Pátria 130, 1169-056 Lisboa, Portugal
| | - Daniela F Santos
- CEDOC - Chronic Diseases Center, NOVA Medical School, Universidade Nova de Lisboa, Campo Mártires da Pátria 130, 1169-056 Lisboa, Portugal; ProRegeM - PhD Programme Mechanisms of Disease and Regenerative Medicine, NOVA Medical School, Universidade NOVA de Lisboa, Campo Mártires da Pátria 130, 1169-056 Lisboa, Portugal
| | - Gabriela A Silva
- CEDOC - Chronic Diseases Center, NOVA Medical School, Universidade Nova de Lisboa, Campo Mártires da Pátria 130, 1169-056 Lisboa, Portugal.
| |
Collapse
|
4
|
Abstract
Neural stem cells (NSCs) have been proposed as a promising cellular source for the treatment of diseases in nervous systems. NSCs can self-renew and generate major cell types of the mammalian central nervous system throughout adulthood. NSCs exist not only in the embryo, but also in the adult brain neurogenic region: the subventricular zone (SVZ) of the lateral ventricle. Embryonic stem (ES) cells acquire NSC identity with a default mechanism. Under the regulations of leukemia inhibitory factor (LIF) and fibroblast growth factors, the NSCs then become neural progenitors. Neurotrophic and differentiation factors that regulate gene expression for controlling neural cell fate and function determine the differentiation of neural progenitors in the developing mammalian brain. For clinical application of NSCs in neurodegenerative disorders and damaged neurons, there are several critical problems that remain to be resolved: 1) how to obtain enough NSCs from reliable sources for autologous transplantation; 2) how to regulate neural plasticity of different adult stem cells; 3) how to control differentiation of NSCs in the adult nervous system. In order to understand the mechanisms that control NSC differentiation and behavior, we review the ontogeny of NSCs and other stem cell plasticity of neuronal differentiation. The role of NSCs and their regulation by neurotrophic factors in CNS development are also reviewed.
Collapse
Affiliation(s)
- Yi-Chao Hsu
- Stem Cell Research Center, National Health Research Institutes, Jhunan, Taiwan
| | - Don-Ching Lee
- Stem Cell Research Center, National Health Research Institutes, Jhunan, Taiwan
| | - Ing-Ming Chiu
- Stem Cell Research Center, National Health Research Institutes, Jhunan, Taiwan
- Department of Internal Medicine, Ohio State University, Columbus, OH 43210, USA
- Institute of Medical Technology, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
5
|
Förthmann B, Grothe C, Claus P. A nuclear odyssey: fibroblast growth factor-2 (FGF-2) as a regulator of nuclear homeostasis in the nervous system. Cell Mol Life Sci 2015; 72:1651-62. [PMID: 25552245 PMCID: PMC11113852 DOI: 10.1007/s00018-014-1818-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 12/10/2014] [Accepted: 12/19/2014] [Indexed: 01/07/2023]
Abstract
Nuclear localization of classical growth factors is a well-known phenomenon but still remains a molecular and cellular conundrum. Fibroblast growth factor-2 (FGF-2) is an excellent example of a protein which functions as an extracellular molecule involved in canonical receptor tyrosine kinase signaling as well as displaying intracellular functions. Paracrine and nuclear functions are two important sides of the same protein. FGF-2 is expressed in isoforms with different molecular weights from one mRNA species. In rodents, all of these isoforms become imported to the nucleus. In this review, we discuss structural and functional aspects of FGF-2 isoforms in the nervous system. The nuclear odyssey of FGF-2 is reflected by nuclear dynamics, localization to nuclear bodies such as nucleoli, binding to chromatin and engagement in various protein interactions. Recently discovered molecular partnerships of the isoforms shed light on their nuclear functions, thereby greatly extending our knowledge of the multifaceted functions of FGF-2.
Collapse
Affiliation(s)
- Benjamin Förthmann
- Department of Neuroanatomy, Institute of Neuroanatomy, Hannover Medical School, OE 4140, Carl-Neuberg-Str.1, 30625 Hannover, Germany
| | - Claudia Grothe
- Department of Neuroanatomy, Institute of Neuroanatomy, Hannover Medical School, OE 4140, Carl-Neuberg-Str.1, 30625 Hannover, Germany
- Center for Systems Neuroscience, 30625 Hannover, Germany
| | - Peter Claus
- Department of Neuroanatomy, Institute of Neuroanatomy, Hannover Medical School, OE 4140, Carl-Neuberg-Str.1, 30625 Hannover, Germany
- Center for Systems Neuroscience, 30625 Hannover, Germany
| |
Collapse
|
6
|
Lauzon MA, Daviau A, Marcos B, Faucheux N. Growth factor treatment to overcome Alzheimer's dysfunctional signaling. Cell Signal 2015; 27:1025-38. [PMID: 25744541 DOI: 10.1016/j.cellsig.2015.02.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 02/16/2015] [Indexed: 10/23/2022]
Abstract
The number of people suffering from Alzheimer's disease (AD) will increase as the world population ages, creating a huge socio-economic burden. The three pathophysiological hallmarks of AD are the cholinergic system dysfunction, the β-amyloid peptide deposition and the Tau protein hyperphosphorylation. Current treatments have only transient effects and each tends to concentrate on a single pathophysiological aspect of AD. This review first provides an overall view of AD in terms of its pathophysiological symptoms and signaling dysfunction. We then examine the therapeutic potential of growth factors (GFs) by showing how they can overcome the dysfunctional cell signaling that occurs in AD. Finally, we discuss new alternatives to GFs that help overcome the problem of brain uptake, such as small peptides, with evidence from some of our unpublished data on human neuronal cell line.
Collapse
Affiliation(s)
- Marc-Antoine Lauzon
- Cell-Biomaterial Biohybrid Systems, Department of Chemical and Biotechnological Engineering, Université de Sherbrooke, 2500 boul. de l'Université, Sherbrooke, Québec J1K 2R1, Canada
| | - Alex Daviau
- Cell-Biomaterial Biohybrid Systems, Department of Chemical and Biotechnological Engineering, Université de Sherbrooke, 2500 boul. de l'Université, Sherbrooke, Québec J1K 2R1, Canada
| | - Bernard Marcos
- Department of Chemical and Biotechnological Engineering, Université de Sherbrooke, 2500 boul. de l'Université, Sherbrooke, Québec J1K 2R1, Canada
| | - Nathalie Faucheux
- Cell-Biomaterial Biohybrid Systems, Department of Chemical and Biotechnological Engineering, Université de Sherbrooke, 2500 boul. de l'Université, Sherbrooke, Québec J1K 2R1, Canada.
| |
Collapse
|
7
|
Kay P, Yang YC, Paraoan L. Directional protein secretion by the retinal pigment epithelium: roles in retinal health and the development of age-related macular degeneration. J Cell Mol Med 2013; 17:833-43. [PMID: 23663427 PMCID: PMC3822888 DOI: 10.1111/jcmm.12070] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Accepted: 03/24/2013] [Indexed: 11/29/2022] Open
Abstract
The structural and functional integrity of the retinal pigment epithelium (RPE) is fundamental for maintaining the function of the neuroretina. These specialized cells form a polarized monolayer that acts as the retinal–blood barrier, separating two distinct environments with highly specialized functions: photoreceptors of the neuroretina at the apical side and Bruch's membrane/highly vascularized choriocapillaris at the basal side. The polarized nature of the RPE is essential for the health of these two regions, not only in nutrient and waste transport but also in the synthesis and directional secretion of proteins required in maintaining retinal homoeostasis and function. Although multiple malfunctions within the RPE cells have been associated with development of age-related macular degeneration (AMD), the leading cause of legal blindness, clear causative processes have not yet been conclusively characterized at the molecular and cellular level. This article focuses on the involvement of directionally secreted RPE proteins in normal functioning of the retina and on the potential association of incorrect RPE protein secretion with development of AMD. Understanding the importance of RPE polarity and the correct secretion of essential structural and regulatory components emerge as critical factors for the development of novel therapeutic strategies targeting AMD.
Collapse
Affiliation(s)
- Paul Kay
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | | | | |
Collapse
|
8
|
Blüm T, Hoheisel U, Unger T, Mense S. Fibroblast growth factor-2 acutely influences the impulse activity of rat dorsal horn neurones. Neurosci Res 2001; 40:115-23. [PMID: 11377749 DOI: 10.1016/s0168-0102(01)00217-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The neurotrophic and neuroprotective actions of fibroblast growth factor-2 (FGF-2) are well-established. The signal cascade mediating these effects includes steps that are likely to influence also the electrical properties of neurones. However, the possibility that FGF-2 may acutely affect the processing of neuronal impulse activity is largely unexplored. In the present study the impulse activity of single dorsal horn neurones was recorded in the rat during ionophoretical administration of FGF-2 close to the neurones. Before and during FGF-2 ionophoresis the receptive field of each cell was tested with defined mechanical stimuli. At a concentration of 10 nM in the ionophoresis pipette, FGF-2 reduced the responses of the cells to mechanical stimulation. There was no preferential action of FGF-2 on a particular functional type of dorsal horn neurone; both non-nociceptive and nociceptive cells exhibited a reduced mechanical responsiveness. The background (ongoing) activity was also depressed in most neurones. The results of the study show that in addition to neurotrophic and neuroprotective actions FGF-2 has an acute inhibitory influence on the impulse activity of spinal sensory neurones. This depression of neuronal activity could add to the neuroprotective action of FGF-2 by counteracting glutamate excitotoxicity following a central nervous trauma.
Collapse
Affiliation(s)
- T Blüm
- Institut für Anatomie und Zellbiologie, Im Neuenheimer Feld 307, D-69120, Heidelberg, Germany
| | | | | | | |
Collapse
|
9
|
Abstract
Although the neuropathological changes caused by severe or repeated seizures have been well characterized, many questions about the molecular mechanisms involved remain unanswered. Neuronal cell death, reactive gliosis, enhanced neurogenesis, and axonal sprouting are four of the best-studied sequelae of seizures. In vitro, each of these pathological processes can be substantially influenced by soluble protein factors, including neurotrophins, cytokines, and growth factors. Furthermore, many of these proteins and their receptors are expressed in the adult brain and are up-regulated in response to neuronal activity and injury. We review the evidence that these intercellular signaling proteins regulate seizure activity as well as subsequent pathology in vivo. As nerve growth factor and brain derived neurotrophic factor are the best-studied proteins of this class, we begin by discussing the evidence linking these neurotrophins to epilepsy and seizure. More than a dozen additional cytokines, growth factors, and neurotrophins that have been examined in the context of epilepsy models are then considered. We discuss the effect of seizure on expression of cytokines and growth factors, and explore the regulation of seizure development and aftermath by exogenous application or antagonist perturbation of these proteins. The experimental evidence supports a role for these factors in each aspect of seizure and pathology, and suggests potential targets for future therapeutics.
Collapse
Affiliation(s)
- J L Jankowsky
- Biology Division, California Institute of Technology, 216-76 Caltech, Pasadena, CA 91125, USA
| | | |
Collapse
|
10
|
Désiré L, Courtois Y, Jeanny JC. Endogenous and exogenous fibroblast growth factor 2 support survival of chick retinal neurons by control of neuronal neuronal bcl-x(L) and bcl-2 expression through a fibroblast berowth factor receptor 1- and ERK-dependent pathway. J Neurochem 2000; 75:151-63. [PMID: 10854258 DOI: 10.1046/j.1471-4159.2000.0750151.x] [Citation(s) in RCA: 68] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Fibroblast growth factor (FGF) 2 is a survival factor for various cell types, including retinal neurons. However, little is understood about the molecular bases of the neuroprotective role of FGF2 in the retina. In this report, FGF2 survival activity was studied in chick retinal neurons subjected to apoptosis by serum deprivation. Exogenous FGF2 supported neuronal survival after serum deprivation and increased neuronal bcl-x(L) and bcl-2 expression, through binding to its receptor R1 (FGF-R1), and subsequent extracellular signal-regulated kinase (ERK) activation. Endogenous FGF2 was transiently overexpressed after serum deprivation. Its down-regulation by antisense oligonucleotides and blockade of its signaling pathway (binding to FGF-R1, tyrosine phosphorylation, and ERK inhibition) decreased bcl-x(L) and bcl-2 levels and and enhanced apoptosis, suggesting that endogenous FGF2 supported neuronal survival through a pathway similar to that of exogenous FGF2. This pathway may serve to up-regulate, or maintain, bcl-x(L) and bcl-2 levels that normally decrease during the onset of apoptosis. Indeed, long-term ERK activation and high bcl-x(L) levels are necessary for the survival activity of both exogenous and endogenous FGF2. Because FGF2 is upregulated following retinal injury in vivo, we suggest that an injury-stimulated autocrine/paracrine FGF2 loop may serve to maintain high levels of survival proteins, such as Bcl-x(L), through ERK activation in retinal neurons.
Collapse
Affiliation(s)
- L Désiré
- INSERM U. 450, Développement, Vieillissement et Pathologie de la Rétine, INSERM, Affiliée CNRS, Association Claude Bernard, Paris, France
| | | | | |
Collapse
|
11
|
Gallo F, Morale MC, Spina-Purrello V, Tirolo C, Testa N, Farinella Z, Avola R, Beaudet A, Marchetti B. Basic fibroblast growth factor (bFGF) acts on both neurons and glia to mediate the neurotrophic effects of astrocytes on LHRH neurons in culture. Synapse 2000; 36:233-53. [PMID: 10819902 DOI: 10.1002/(sici)1098-2396(20000615)36:4<233::aid-syn1>3.0.co;2-i] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Luteinizing hormone-releasing hormone (LHRH) neurons play a pivotal role in the neuroendocrine control of mammalian reproduction. Astrocytes were shown to be involved in the regulation of LHRH neuronal function, but little is known about the contribution of astroglial-derived factors in the regulation of LHRH neuron development. In order to gain insight into the mechanisms regulating the development of these cells, at morphological and biochemical levels we characterized the neurotrophic effects exerted by young astrocytes (maintained in culture for 8 days in vitro) and old astrocytes (maintained 26 days) on the differentiation, proliferation, and phenotypic expression of immortalized hypothalamic LHRH (GT(1-1)) neurons in vitro. Culturing GT(1-1) cells in the presence of young glia for different time intervals caused a marked acceleration in the acquisition of their neuronal phenotype. At all times examined, GT(1-1) cells cocultured with young glia exhibited a significantly greater extension of processes/cell, larger number of processes/cell and greater surface area of growth cones than GT(1-1) cells grown over nonglial adhesive substrates (polylysine). By contrast, when GT(1-1) neurons were cocultured with old glia, the length of neuronal processes and the growth cone surface area were significantly lower than in control GT(1-1) neurons cultured in the absence of glia. At 3 days in vitro (DIV), GT(1-1) neurons cocultured with young glia exhibited a 50% lower incorporation of [(3)H]thymidine than GT(1-1) neurons cultured without glia. By contrast, in the presence of old glia [(3)H]thymidine incorporation was significantly higher in cells cocultured with glia than in GT(1-1) neurons cultured alone. Localization of the proliferating cells by dual immunohistochemical staining revealed that the incorporation of bromodeoxiuridine (BrdU) was restricted to nuclei of GT(1-1) neurons when these were cocultured with young glia, but associated with both neurons and astrocytes in the presence of old glia. At the functional level, coculture of GT(1-1) neurons with young glia increased the spontaneous release of LHRH as compared to GT(1-1) neurons grown in the absence of glia. By contrast, in the presence of old glia LHRH release in the medium was significantly lower than in controls. Conditioned medium of young glia (ACM-Y) induced significant neurotrophic and functional effects on GT(1-1) cells, but these effects were 50% less potent than the coculture itself. Heat denaturation of ACM-Y totally abolished its neurotrophic and functional properties, indicating that they involved a peptide factor. Suppression of bFGF activity in ACM-Y reduced its neurotrophic activity by approximately 40%, but did not affect its LHRH release-promoting effects. By contrast, neutralization of endogenous bFGF activity in GT(1-1) neurons cocultured with young glia counteracted both neurotrophic and functional effects of young glia. Treatment of old glia with bFGF rescued its neurotrophic and functional effects on GT(1-1) cells. Moreover, the ACM of aged bFGF-treated old glia was the most powerful neurotrophic stimulus for GT(1-1) neurons. These results suggest that: 1) soluble peptidic factors, including bFGF, and mechanism(s) requiring coculture are responsible for the highly potent neurotrophic and functional effects of young glia; 2) the inhibitory effects of old glia on neurite outgrowth and LHRH release are mediated in part by soluble inhibitory molecules and in part by factors requiring coculture with old glia; 3) old glia may revert to a growth-supporting state when treated with bFGF and this functional shift involves a diffusible molecule with potent neurotrophic and functional effects on immortalized LHRH neurons. (c) 2000 Wiley-Liss, Inc.
Collapse
Affiliation(s)
- F Gallo
- Department of Pharmacology, Medical School, University of Catania, 95125 Catania, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
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: 1.2] [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
|
13
|
Naffah-Mazzacoratti MG, Argañaraz GA, Porcionatto MA, Scorza FA, Amado D, Silva R, Bellissimo MI, Nader HB, Cavalheiro EA. Selective alterations of glycosaminoglycans synthesis and proteoglycan expression in rat cortex and hippocampus in pilocarpine-induced epilepsy. Brain Res Bull 1999; 50:229-39. [PMID: 10582521 DOI: 10.1016/s0361-9230(99)00195-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Proteoglycans and glycosaminoglycans are elements of matrix. In the nervous system, glycosaminoglycans modulate neurite outgrowth and are co-receptors for growth factors playing a crucial role in cell differentiation and synaptogenesis. The receptor of protein tyrosine phosphatase beta (RPTPbeta) is a chondroitin sulphate proteoglycan which plays an important role in neural morphogenesis and axon guidance mechanisms. Pilocarpine-treated rats present status epilepticus, which is followed by a seizure-free period (silent), by a period of spontaneous recurrent seizures (chronic), and the hippocampus of these animals exhibits cell loss and mossy fiber sprouting. Thus, the synthesis of heparan sulphate and chondroitin sulphate and the time course of RPTPbeta immunoreactivity were studied in the hippocampus and cerebral cortex during these phases of pilocarpine-induced epilepsy. The results showed decreased synthesis of heparan sulphate during the acute phase and an increased synthesis of chondroitin sulphate during the silent period in the cortex and hippocampus. In control rats RPTPbeta immunoreactivity was detected only in glial cells. After 6 h of status epilepticus the RPTPbeta immunoreactivity was no longer detectable in the glial cells in both tissues and intense staining became evident in the matrix, surrounding CA3 and dentate gyrus and piriform cortex neurones. In the silent and chronic periods RPTPbeta immunoreactivity was mainly detected in neuronal somata and fibers of neurones of hippocampus and cortex. These changes show a selective variation of synthesis and expression of glycosaminoglycans and RPTPbeta in relation to epilepsy suggesting a molecular interplay between glia and neurones during seizures.
Collapse
|
14
|
Raguenez G, Desire L, Lantrua V, Courtois Y. BCL-2 is upregulated in human SH-SY5Y neuroblastoma cells differentiated by overexpression of fibroblast growth factor 1. Biochem Biophys Res Commun 1999; 258:745-51. [PMID: 10329457 DOI: 10.1006/bbrc.1999.0613] [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: 11/22/2022]
Abstract
Fibroblast growth factor 1 (FGF1) is a multipotent factor in the development and differentiation of the central nervous system. Recent studies in PC12 cells attribute these effects to high endogenous FGF1 expression. To examine the differentiation mechanisms induced by FGF1, we performed studies in SH-SY5Y human neuroblastoma cells. We monitored the impact of FGF1 overexpression in SH-SY5Y either after addition of exogenous FGF1 and heparin or after stable transfection with the FGF1 eukaryotic expression vector. Under both conditions, the FGF1 endogenous rise caused SH-SY5Y cell differentiation with morphological changes (appearance of neuritic extensions), increased GAP-43 gene expression, decreased of N-myc gene expression, and prolonged long-term survival in serum-free media. These modifications were correlated with Bcl-2 upregulation. These results suggest that there is a link between the endogenous FGF1 signaling pathway and Bcl-2 in neuronal survival modulation.
Collapse
Affiliation(s)
- G Raguenez
- Développement, Vieillissement et Pathologie de la Rétine, INSERM U. 450, Affiliée CNRS, Association Claude Bernard - 29 rue Wilhem, Paris, 75016, France.
| | | | | | | |
Collapse
|
15
|
Charon I, Zuin-Kornmann G, Bataillé S, Schorderet M. Protective effect of neurotrophic factors, neuropoietic cytokines and dibutyryl cyclic AMP on hydrogen peroxide-induced cytotoxicity on PC12 cells: a possible link with the state of differentiation. Neurochem Int 1998; 33:503-11. [PMID: 10098719 DOI: 10.1016/s0197-0186(98)00056-4] [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: 10/17/2022]
Abstract
We present evidence that the survival of PC12 cells exposed to hydroxyl radicals generated by hydrogen peroxide applied for 30 min at 1 mM was effective when they were differentiated in response to Nerve Growth Factor (NGF) and/or other inducers of neurite outgrowth such as basic-fibroblast growth factor and dibutyryl cyclic AMP. The time- and dose-dependent differentiation triggered by NGF was (1) markedly increased by basic fibroblast growth factor, interleukin-6 or dibutyryl cyclic AMP; (2) diminished by leukemia inhibitory factor or ciliary neurotrophic factor; (3) not potentiated by insulin-like growth factor I or progesterone. The influence of these various factors and agents on PC12 cells was evaluated by the estimation of neurite outgrowth, whereas their possible protective effects were assessed by the measurement of cell survival. Our results would indicate that the factors and agents that induced differentiation were also able to protect the cells against an oxidative stress.
Collapse
Affiliation(s)
- I Charon
- Department of Pharmacology, University Medical Center, Geneva, Switzerland
| | | | | | | |
Collapse
|
16
|
Szebenyi G, Fallon JF. Fibroblast growth factors as multifunctional signaling factors. INTERNATIONAL REVIEW OF CYTOLOGY 1998; 185:45-106. [PMID: 9750265 DOI: 10.1016/s0074-7696(08)60149-7] [Citation(s) in RCA: 356] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The fibroblast growth factor (FGF) family consists of at least 15 structurally related polypeptide growth factors. Their expression is controlled at the levels of transcription, mRNA stability, and translation. The bioavailability of FGFs is further modulated by posttranslational processing and regulated protein trafficking. FGFs bind to receptor tyrosine kinases (FGFRs), heparan sulfate proteoglycans (HSPG), and a cysteine-rich FGF receptor (CFR). FGFRs are required for most biological activities of FGFs. HSPGs alter FGF-FGFR interactions and CFR participates in FGF intracellular transport. FGF signaling pathways are intricate and are intertwined with insulin-like growth factor, transforming growth factor-beta, bone morphogenetic protein, and vertebrate homologs of Drosophila wingless activated pathways. FGFs are major regulators of embryonic development: They influence the formation of the primary body axis, neural axis, limbs, and other structures. The activities of FGFs depend on their coordination of fundamental cellular functions, such as survival, replication, differentiation, adhesion, and motility, through effects on gene expression and the cytoskeleton.
Collapse
Affiliation(s)
- G Szebenyi
- Anatomy Department, University of Wisconsin, Madison 53706, USA
| | | |
Collapse
|
17
|
Milev P, Monnerie H, Popp S, Margolis RK, Margolis RU. The core protein of the chondroitin sulfate proteoglycan phosphacan is a high-affinity ligand of fibroblast growth factor-2 and potentiates its mitogenic activity. J Biol Chem 1998; 273:21439-42. [PMID: 9705269 DOI: 10.1074/jbc.273.34.21439] [Citation(s) in RCA: 76] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Using a radioligand binding assay we have demonstrated that phosphacan, a chondroitin sulfate proteoglycan of nervous tissue that also represents the extracellular domain of a receptor-type protein tyrosine phosphatase, shows saturable, reversible, high-affinity binding (Kd approximately 6 nM) to fibroblast growth factor-2 (FGF-2). Binding was reduced by only approximately 35% following chondroitinase treatment of the proteoglycan, indicating that the interaction is mediated primarily through the core protein rather than the glycosaminoglycan chains. Immunocytochemical studies also showed an overlapping localization of FGF-2 and phosphacan in the developing central nervous system. At concentrations of 10 microg protein/ml, both native phosphacan and the core protein obtained by chondroitinase treatment potentiated the mitogenic effect of FGF-2 (5 ng/ml) on NIH/3T3 cells by 75-90%, which is nearly the same potentiation as that produced by heparin at an equivalent concentration. Although studies on the role of proteoglycans in mediating the binding and mitogenic effects of FGF-2 have previously focused on cell surface heparan sulfate, our results indicate that the core protein of a chondroitin sulfate proteoglycan may also regulate the access of FGF-2 to cell surface signaling receptors in nervous tissue.
Collapse
Affiliation(s)
- P Milev
- Department of Pharmacology, New York University Medical Center, New York, New York 10016, USA
| | | | | | | | | |
Collapse
|
18
|
Désiré L, Courtois Y, Jeanny JC. Suppression of fibroblast growth factors 1 and 2 by antisense oligonucleotides in embryonic chick retinal cells in vitro inhibits neuronal differentiation and survival. Exp Cell Res 1998; 241:210-21. [PMID: 9633530 DOI: 10.1006/excr.1998.4048] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
As retinal histogenesis proceeds there is a pronounced increase in the expression of fibroblast growth factor (FGF), reaching its maximum in the mature retina and largely in terminal differentiated retinal neurons. Recent in vivo evidence suggests that exogenous FGF functions as a differentiation and survival factor for a wide variety of cell types including CNS neurons and that endogenous FGF may perform similar functions. We have examined the consequences of selectively and independently inhibiting FGF1 or FGF2 expression using antisense oligonucleotides in embryonic chick retinal cells, differentiating in vitro. Whether FGF1 or FGF2 expression was inhibited the results were the same: a marked reduction in neuronal photoreceptor cells differentiation, an increase in programmed cell death, but no effects on cell proliferation. Even although these two related factors promote the same final effect on retinal cells, namely, neuronal differentiation and survival, their normal combined activities or levels appear to be important in achieving this effect. Stimulation with either exogenous FGF1 or FGF2 served to increase endogenous levels of both FGF1 and FGF2 and reversed the effects of antisense blockade of either FGF1 or FGF2. Our data suggest that although other sources of FGF exist within the eye, the function of endogenous FGF in differentiating retinal neurons may be to stimulate their differentiation and promote their survival.
Collapse
Affiliation(s)
- L Désiré
- Développement, vieillissement et pathologie de la rétine, INSERM U. 450, affiliée CNRS, Paris, France
| | | | | |
Collapse
|
19
|
Désiré L, Head MW, Fayein NA, Courtois Y, Jeanny JC. Suppression of fibroblast growth factor 2 expression by antisense oligonucleotides inhibits embryonic chick neural retina cell differentiation and survival in vivo. Dev Dyn 1998; 212:63-74. [PMID: 9603424 DOI: 10.1002/(sici)1097-0177(199805)212:1<63::aid-aja6>3.0.co;2-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
During retinal differentiation, fibroblast growth factor 2 (FGF2) expression increases in retinal neurons following the sequential appearance of the neuronal layers. The function of the developmental increase of endogenous FGF2 in the developing chick retina was investigated by using an antisense strategy, using both optic vesicle cultures and in ovo-intravitreal microinjections. The former model allowed us to study the consequences of FGF2 down-regulation on early ganglion cell differentiation, whereas, in the latter model, subsequent development stages and terminal maturation of the retina were studied. FGF2 inhibition resulted in reduced ganglion cell differentiation, as visualized by the expression of the ganglion cell-specific RA4 and Islet-1 markers in optic vesicle cultures. Eyes intravitreally injected with the FGF2-specific antisense oligonucleotide exhibited profound retinal differentiation defects: thinning of the ganglion and outer nuclear (photoreceptors) cell layers and increased cell death in ganglion cell and inner nuclear layers. These results indicate that the loss of endogenous FGF2 cannot be compensated for in the retina and suggest that, although many other sources of FGF exist in the eye, the main role of the increase in endogenous FGF2 observed during retinal development is to intrinsically stimulate neuron differentiation and to protect neurons against cell death.
Collapse
Affiliation(s)
- L Désiré
- Développement, Vieillissement et Pathologie de la Rétine, Institut National de la Santé et de la Recherche Médicale, Affiliée CNRS, Association Claude Bernard, Paris, France
| | | | | | | | | |
Collapse
|
20
|
Cuevas P, Carceller F, Nieto I, Giménez-Gallego G. Spasmolytic effect of acidic fibroblast growth factor in early cerebral vasospasm in the rat. SURGICAL NEUROLOGY 1998; 49:176-9; discussion 179-80. [PMID: 9457268 DOI: 10.1016/s0090-3019(97)00167-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND In animal models of subarachnoid hemorrhage (SAH), basilar artery spasm has been described. Since fibroblast growth factors (FGFs) dilate rat basilar artery in vivo, we tested whether systemic administration of acidic fibroblast growth factor (aFGF) can reverse the basilar artery spasm following rat SAH. METHODS SAH was mimicked by injecting autologous blood into the cisterna magna. We used a digital substraction angiography system in order to evaluate the spasmolytic effect of aFGF administered via the axillary artery 7 minutes after the injection of the autologous blood. Control animals received vehicle solution in the same manner. RESULTS Intraarterial bolus injection of 2.6 micrograms aFGF, but not the solvent, reverses the acute basilar artery narrowing caused by SAH (23.05% of the baseline value at 5 minutes post-SAH). CONCLUSIONS Our results suggest an important role for FGFs in the cerebral tone regulation and support a clinical interest of FGFs in preventing cerebral ischemia following SAH, particularly if these vasoactive effects are added to the known neuroprotective effects of FGFs.
Collapse
Affiliation(s)
- P Cuevas
- Servicio de Histología, Hospital Ramón y Cajal, Madrid, Spain
| | | | | | | |
Collapse
|
21
|
Ferrer I, Martí E. Distribution of fibroblast growth factor receptor-1 (FGFR-1) and FGFR-3 in the hippocampus of patients with Alzheimer's disease. Neurosci Lett 1998; 240:139-42. [PMID: 9502223 DOI: 10.1016/s0304-3940(97)00948-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
To learn about the localization of fibroblast growth factor (FGF) ligands in normal and pathologic brains, fibroblast growth factor receptor-1 (FGFR-1; Flg) and FGFR-3 immunoreactivities were examined in the hippocampus of patients with Alzheimer's disease and age-matched controls. Flg immunoreactivity was found in practically all neurons of the hippocampus and dentate gyrus in control and Alzheimer's disease cases. In patients with Alzheimer's disease, Flg immunoreactivity was present in tangle-bearing and non-tangle-bearing neurons, as well as in neurons with granulovacuolar degeneration, but not in ghost tangles. Aberrant neurites of senile plaques were negative. FGFR-3 immunoreactivity was found in reactive glial cells, most of them astrocytes, including those in the vicinity of senile plaques.
Collapse
Affiliation(s)
- I Ferrer
- Unitat de Neuropatología, Servei d'Anatomía Patològica, Hospital Princeps d'Espanya, Barcelona, Spain
| | | |
Collapse
|
22
|
Abstract
This study describes the spatio-temporal expression of basic Fibroblast growth factor (FGF-2) during odontogenesis of mouse as revealed by immunohistology. Parasagittal sections of mouse embryo head (13-18 day of gestation) containing various stages of developing tooth were incubated with a polyclonal anti-FGF-2 antibody and positive binding was evidentiated by using Streptavidin-Biotin complex-HRP system and AEC staining. We observed no FGF-2 staining at the dental lamina stage. At the bud stage slight staining is seen, limited to some epithelial cells. The intensity of the staining increases at the cap stage. In the bell stage, the stellate reticulum cells stain intensely. Later, odontoblasts and the dentin matrix stain deeply; but the epithelial cells stain faint. The extra cellular matrix of the dentin and dental papilla stain very intense but the enamel matrix is found negative. These results indicate the participation of FGF-2 in differentiation rather than in proliferation of tooth-forming cells. In particular, it appears that FGF-2 participates in odontoblast differentiation and in dentin matrix deposition. The spatio-temporally specific distribution pattern of FGF-2 in developing mouse tooth reported here emphasizes the importance of FGF-2 in mammalian odontogenesis.
Collapse
Affiliation(s)
- L G Russo
- Department of Structure, Function and Biotechnology, Faculty of Veterinary Medicine, University of Naples Federico II, Italy
| | | | | |
Collapse
|
23
|
Abstract
Fibroblast growth factor (FGF)-2 differentially regulates oligodendrocyte progenitor proliferation and differentiation in culture, and modulates gene expression of its own receptors, in a developmental and receptor type-specific manner (Bansal et al., 1996a,b). Three FGF receptors (types 1, 2, 3) are expressed in postmitotic, terminally differentiating oligodendrocytes. Exposure of such cells to FGF-2 results in: (a) the down-regulation of myelin-specific gene expression (e.g., ceramide galactosyltransferase, 2',3'-cyclic nucleotide 3'-phosphohydrolase, myelin basic protein, proteolipid protein), (b) dramatic increases in the length of cellular processes in a time- and dose-dependent manner, (c) re-entrance into the cell cycle without accompanying mitosis, and (d) the alteration of the expression of both low- and high-affinity FGF receptors. Compared to oligodendrocyte progenitors, the differentiated oligodendrocytes treated with FGF-2 incorporate BrdU at a slower rates, exhibit different patterns of both FGF high- and low-affinity (syndecans) receptors, and are morphologically very different. In addition, they do not re-express the progenitor markers A2B5, NG2 or PDGFalpha receptor. Therefore, although the FGF-treated cells lose their differentiated OL/myelin markers, they do not revert to progenitors and clearly represent a different, apparently novel, phenotype both morphologically and biochemically, which we have termed NOLs. These data indicate that terminally differentiated oligodendrocytes retain the plasticity to reprogram their differentiation fate under the influence of environmental factors. The possible significance of this response to FGF relative to normal and pathological physiology is discussed. In particular, on the basis of these data we predict the appearance of cells in and around multiple sclerosis plaques with the phenotype O4+, NG2-, A2B5-, O1-, MBP-.
Collapse
Affiliation(s)
- R Bansal
- Department of Pharmacology, University of Connecticut Medical School, Farmington 06030-3205, USA.
| | | |
Collapse
|
24
|
Blottner D, Stapf C, Meisinger C, Grothe C. Localization, differential expression and retrograde axonal transport suggest physiological role of FGF-2 in spinal autonomic neurons of the rat. Eur J Neurosci 1997; 9:368-77. [PMID: 9058056 DOI: 10.1111/j.1460-9568.1997.tb01406.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Fibroblast growth factor-2 (FGF-2) has marked pharmacological neurotrophic effects on lesioned spinal autonomic neurons following target removal of the adrenal medulla, yet expression and axonal transport in autonomic neurons remain to be shown. We show here FGF-2 and FGF receptor type 1 (FGFR1) protein and mRNA expression in preganglionic intermediolateral neurons of the rat thoracic spinal cord. While immunoreactivity of both FGF-2 and FGFR1 co-localize to intermediolateral neurons, mRNA transcripts of FGFR1, but not of FGF-2, are detectable in intermediolateral preparations by RNase protection analysis, suggesting protein translocation in vivo. Unilateral microinjection of 125iodinated FGF-2 into the adrenal medulla (a major target of intermediolateral neurons) results in significant accumulation of specific radioactivity in thoracic spinal cord tissue, including the intermediolateral neurons, and the ipsilateral splanchnic nerve. Emulsion autoradiography demonstrated labelling over ipsilateral intermediolateral neurons only. Neuronal co-localization of FGF-2/FGFR1 protein, differential mRNA expression, specific retrograde axonal transport and the known neurotrophic actions in vivo, strongly suggest unique physiological roles of FGF-2 in the autonomic nervous system.
Collapse
Affiliation(s)
- D Blottner
- Institute for Anatomy, University Clinics Benjamin Franklin, Freie Universität Berlin, Germany
| | | | | | | |
Collapse
|
25
|
Zhou X, Hossain WA, Rutledge A, Baier C, Morest DK. Basic fibroblast growth factor (FGF-2) affects development of acoustico-vestibular neurons in the chick embryo brain in vitro. Hear Res 1996; 101:187-207. [PMID: 8951444 DOI: 10.1016/s0378-5955(96)00122-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The effects of basic fibroblast growth factor (FGF-2) on presumptive auditory and vestibular neurons from the medulla were studied in primary cell cultures. The part of the rhombic lip that forms nucleus magnocellularis (homologue of the mammalian anteroventral cochlear nucleus) was explanted from white leghorn chicken embryos at Hamburger-Hamilton stage 28 (E5.5), the time when precursors of the magnocellularis bushy cells migrate and begin to differentiate in situ. In vitro the neuroblasts migrated onto 2-D substrates of purified collagen, differentiated, and expressed neuronal markers. One-half of the cultures were supplemented with human recombinant FGF-2 (10 ng/ml daily) for 5-7 days; the others, with fetal bovine serum. FGF-2 more than doubled the length of neurite outgrowth during the first 3 day treatment compared to serum, but the number of migrating neuroblasts was unaffected. Although neurites attained greater lengths in FGF-2, they usually degenerated after 4-5 days; in serum their growth continued for several weeks. Differentiation of neuronal structure, including axons and dendrites, began within 1-2 days in bFGF but required at least 5-7 days in serum. Histochemical observations in vitro and in situ with antibodies to FGF receptor demonstrated immunopositive patches on acoustico-vestibular neuroblasts at stage 28, when they are migrating and first forming their axons. The findings suggest that FGF-2 stimulates neurite outgrowth in the cochlear and vestibular nuclei. FGF-2 may accelerate cell death by overstimulating neuroblasts, but other factors are needed to sustain their further development.
Collapse
Affiliation(s)
- X Zhou
- Department of Anatomy, University of Connecticut Health Center, Farmington 06030, USA
| | | | | | | | | |
Collapse
|
26
|
Cuevas Sanchez P. How neurotrophic factors protect brain against excitotoxicity. SURGICAL NEUROLOGY 1996; 46:152-3. [PMID: 8685824 DOI: 10.1016/0090-3019(96)00086-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
27
|
Cuevas P, Prieto R, Saenz de Tejada I, Giménez-Gallego G. Analgesic effects of fibroblast growth factor in the rat. Neurosci Lett 1996; 207:175-8. [PMID: 8728478 DOI: 10.1016/0304-3940(96)12526-x] [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: 02/01/2023]
Abstract
Intraperitoneal (i.p.) injection of acidic fibroblast growth factor (aFGF) to Sprague-Dawley rats induced short-lasting analgesia as measured by tail-flick latency (TFL) test. The maximum effect, a 26% increase in tail-flick latency, was obtained 15 min following 1 microgram i.p. aFGF. By 30 min the effect was considerably reduced, and was no longer present by 45 min after treatment. Administration of heat-inactivated aFGF or a hybrid form of aFGF (CLYT/aFGF) that, although active, is unable to cross the blood-brain barrier (BBB), caused no analgesia. Furthermore, the analgesic effects of aFGF were prevented by pretreatment with the nitric oxide synthase inhibitor, L-NG-nitroarginine methyl ester (L-NAME). Our findings demonstrate an analgesic effect of FGF, which requires crossing of BBB and implicates the nitric oxide pathway.
Collapse
Affiliation(s)
- P Cuevas
- Department of Research, Ramón y Cajal Hospital, Madrid, Spain
| | | | | | | |
Collapse
|
28
|
Zhou X, Hossain WA, Rutledge A, Baier C, Morest DK. Basic fibroblast growth factor (FGF-2) affects development of acoustico-vestibular neurons in the chick embryo brain in vitro. Hear Res 1996; 93:147-66. [PMID: 8735076 DOI: 10.1016/0378-5955(95)00222-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The effects of basic fibroblast growth factor (FGF-2) on presumptive auditory and vestibular neurons from the medulla were studied in primary cell cultures. The part of the rhombic lip that forms nucleus magnocellularis (homologue of the mammalian anteroventral cochlear nucleus) was explanted from white leghorn chicken embryos at Hamburger-Hamilton stage 28 (E5.5), the time when precursors of the magnocellularis bushy cells migrate and begin to differentiate in situ. In vitro the neuroblasts migrated onto 2-D substrates of purified collagen, differentiated, and expressed neuronal markers. One-half of the cultures were supplemented with human recombinant FGF-2 (10 ng/ml daily) for 5-7 days; the others, with fetal bovine serum. FGF-2 more than doubled the length of neurite outgrowth during the first 3 day treatment compared to serum, but the number of migrating neuroblasts was unaffected. Although neurites attained greater lengths in FGF-2, they usually degenerated after 4-5 days; in serum their growth continued for several weeks. Differentiation of neuronal structure, including axons and dendrites, began within 1-2 days in bFGF but required at least 5-7 days in serum. Histochemical observations in vitro and in situ with antibodies to FGF receptor demonstrated immunopositive patches on acoustico-vestibular neuroblasts at stage 28, when they are migrating and first forming their axons. The findings suggest that FGF-2 stimulates neurite outgrowth in the cochlear and vestibular nuclei. FGF-2 may accelerate cell death by overstimulating neuroblasts, but other factors are needed to sustain their further development.
Collapse
Affiliation(s)
- X Zhou
- Department of Anatomy, University of Connecticut Health Center, Farmington 06030, USA
| | | | | | | | | |
Collapse
|
29
|
Fayein NA, Head MW, Jeanny JC, Courtois Y, Fuhrmann G. Expression of the chicken cysteine-rich fibroblast growth factor receptor (CFR) during embryogenesis and retina development. J Neurosci Res 1996; 43:602-12. [PMID: 8833095 DOI: 10.1002/(sici)1097-4547(19960301)43:5<602::aid-jnr10>3.0.co;2-2] [Citation(s) in RCA: 11] [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
The expression of the chicken cysteine-rich fibroblast growth factor receptor (CFR) during organogenesis and specifically during retina formation was studied by Northern blotting and a sensitive in situ hybridization. At days 2 and 4 of embryonic development (E2 and E4), CFR mRNA was present in a wide variety of developing organs; it was abundantly expressed in nervous structures, particularly in the retina. The levels of CFR transcripts were high during the proliferation and the subsequent differentiation phases of retinal neurogenesis, reached a maximum around E11 during the onset of the major period of retinal cell death, and then declined progressively. CFR mRNA was not detected at late stages when the final arrangement of retinal cell layers has been established. In prolonged primary cell cultures of chicken embryo retina, CFR expression showed a similar down-regulation to that seen with increasing age in vivo. It was up-regulated either directly or indirectly by its ligands. The CFR expression pattern in the developing retina was complementary to that of two other fibroblast growth factor (FGF) receptors, namely FGF-R1 and FGF-R2. In regard to a progressive increase in the expression of their ligands during retinal development, we suggest that CFR may have a role distinct from that of the tyrosine kinase FGF receptors during retinogenesis. Finally, the comparison of CFR expression with those of the other high affinity receptors indicates a regulation of the FGF function at the receptor level during neural retina development.
Collapse
Affiliation(s)
- N A Fayein
- Unite de Recherches Gerontologiques, INSERM, Developpement et Senescence Cellulaire, Paris, France
| | | | | | | | | |
Collapse
|
30
|
Gehrmann J, Lannes-Vieira J, Wekerle H. Differential expression of fibroblast growth factor-2 and receptor by glial cells in experimental autoimmune encephalomyelitis (EAE). Glia 1996; 16:93-100. [PMID: 8929896 DOI: 10.1002/(sici)1098-1136(199602)16:2<93::aid-glia1>3.0.co;2-b] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
To assess the expression pattern of basic fibroblast growth factor (FGF-2) and one of its receptors (FGFR-1/flg) during autoimmune inflammation of the CNS, FGF-2, and FGFR1/flg peptide and mRNA levels were examined by immunocytochemistry, by in situ hybridisation and by Northern blot analysis in T cell-mediated EAE of the Lewis rat. In naive control animals as well as in animals injected with non-encephalitogenic, PPD-reactive T lymphocytes, FGF-2 immunoreactivity was low and confined to blood vessels and to a few spinal cord neurons. In rats injected with encephalitogenic, MBP-reactive T lymphocytes, however, FGF-2-immunoreactive cells were detected from day 4 after T cell transfer onward, i.e., from the onset of clinical symptoms. The number of FGF-2 immunoreactive cells was highest between days 6 and 10 after T cell transfer. Increased FGF-2 peptide expression was paralleled by increased FGF-2 mRNA expression on macrophages/microglia in the spinal cord. By 21 days after T cell transfer, i.e. after complete recovery, FGF-2 peptide and mRNA expression had fully subsided. Based on morphological criteria and on double labeling with the macrophage/microglia-binding lectin GSI-B4 two cell types expressed FGF-2: 1) round macrophages within the core, and 2) activated microglia at the edges of white and grey matter perivascular lesions. Paralleling the temporal and spatial expression pattern of FGF-2, FGFR-1/flg immunoreactivity was induced on activated macrophages/microglia but also on reactive astrocytes bordering perivascular inflammatory lesions. In situ hybridisation analysis furthermore showed that macrophages/microglia expressed the FGFR-1/flg mRNA, and that receptor mRNA expression paralleled ligand mRNA expression. Macrophage/microglia-derived FGF-2 could serve two main functions in EAE: 1) regulate microglial activation in an autocrine fashion, and 2) help to target astrocyte-derived insulin-like growth factor-I (IGF-I) to potentially injured oligodendrocytes in demyelination.
Collapse
MESH Headings
- Animals
- Blotting, Northern
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/psychology
- Female
- Fibroblast Growth Factor 2/biosynthesis
- Immunohistochemistry
- In Situ Hybridization
- Myelin Sheath/immunology
- Myelin Sheath/ultrastructure
- Neuroglia/metabolism
- RNA, Messenger/biosynthesis
- Rats
- Rats, Inbred Lew
- Receptors, Fibroblast Growth Factor/biosynthesis
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- J Gehrmann
- Institutes of Clinical Pathology and Neuropathology, University Hospital, Zurich, Switzerland
| | | | | |
Collapse
|
31
|
Cuevas P, Giménez-Gallego G. Antiepileptic effects of acidic fibroblast growth factor examined in kainic acid-mediated seizures in the rat. Neurosci Lett 1996; 203:66-8. [PMID: 8742048 DOI: 10.1016/0304-3940(95)12254-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The possibility that exogenous recombinant human acidic fibroblast growth factor (rhaFGF) may have anticonvulsant properties was investigated in a model of temporal lobe epilepsy in awake rats. We found that after intraperitoneal injection of rhaFGF in kainic acid-treated rats, tonic-clonic convulsions and mortality were decreased by 74% and 77%, respectively. These results are consistent with previous studies showing a neuroprotective effect of FGF against insults to the brain and support a possible therapeutic role for FGF in the treatment of excitotoxic processes.
Collapse
Affiliation(s)
- P Cuevas
- Servicio de Histología, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | | |
Collapse
|
32
|
Regeneration of the Auditory Nerve: The Role of Neurotrophic Factors. CLINICAL ASPECTS OF HEARING 1996. [DOI: 10.1007/978-1-4612-4068-6_3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
33
|
Affiliation(s)
- G M Jonakait
- Department of Biological Sciences, Rutgers University, Newark, New Jersey 07102, USA
| |
Collapse
|
34
|
Affiliation(s)
- S McFarlane
- Department of Biology, University of California, San Diego, La Jolla, CA 92093-0366, USA
| | | |
Collapse
|
35
|
Abstract
With so many neurotrophins and receptors now known, how is our picture of neurotrophism changing? Recent studies on knockout mice have confirmed our expectations of neurotrophin action in neuronal development. A notable exception is the activation of TrkB, on motor neurons, by an unknown ligand. It is also clear that some neurotrophins have diverse activities and influence early developmental stages. There are interesting new data concerning the role of p75, the low affinity neurotrophin receptor, as a modulator of neurotrophin activity. Even more exciting are new studies on glia-derived neurotrophic factor (GDNF) which demonstrate that this growth factor acts as a potential protector of motor neurons and striatal dopaminergic neurons.
Collapse
Affiliation(s)
- M C Birling
- Department of Molecular Neuropathology, SmithKline Beecham Pharmaceuticals, Harlow, Essex, UK. Marie-C-Birling%@INET
| | | |
Collapse
|
36
|
McFarlane S, McNeill L, Holt CE. FGF signaling and target recognition in the developing Xenopus visual system. Neuron 1995; 15:1017-28. [PMID: 7576646 DOI: 10.1016/0896-6273(95)90091-8] [Citation(s) in RCA: 148] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
We report that the growth cones of Xenopus retinal ganglion cells express fibroblast growth factor receptors (FGFRs) and that bFGF stimulates neurite extension from cultured retinal neurons. Furthermore, bFGF is abundant in the developing optic tract but is reduced in the optic tectum. To test whether FGF signaling plays a role in axonal guidance in vivo, bFGF was exogenously applied to the developing optic pathway in "exposed brain" preparations. FGF-treated retinal axons navigate normally through the optic tract, but the majority veer aberrantly at the tectal border and bypass the target. Our results implicate FGF signaling in target recognition and suggest that diminished levels of bFGF in the tectum cause arriving axons to slow their growth.
Collapse
Affiliation(s)
- S McFarlane
- Department of Biology, University of California, San Diego, La Jolla 92093, USA
| | | | | |
Collapse
|
37
|
Brickman YG, Ford MD, Small DH, Bartlett PF, Nurcombe V. Heparan sulfates mediate the binding of basic fibroblast growth factor to a specific receptor on neural precursor cells. J Biol Chem 1995; 270:24941-8. [PMID: 7559620 DOI: 10.1074/jbc.270.42.24941] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Heparan sulfate proteoglycans are thought to be obligatory for receptor binding and subsequent mitogenic activity of basic fibroblast growth factor (FGF-2). In a previous study (Nurcombe V., Ford, M. D., Wildschut, J., Bartlett, P. F. (1993) Science 260, 103-106) we have shown that primary cultures of mouse neuroepithelial cells and a cell line derived from then, 2.3D, secrete a heparan sulfate proteoglycan with a high affinity for FGF-2. In this study, a combination of affinity chromatography and gel chromatography was used to further isolate heparan sulfate side chains with high affinity for FGF-2. These active chains had an average molecular weight of 18,000-20,000. In order to determine whether heparan sulfate chains with specificity for FGF-2 also displayed selectivity for the different FGF receptors, peptides designed to the heparin-binding region of the receptors were used in competitive inhibition studies. The structure of the predicted heparin-binding domain of the FGF receptor 1 was modeled on the basis of its presumed secondary and tertiary structure homology with immunoglobulin loops. These results suggested that many of the basic residues within the second immunoglobulin loop of the FGF receptor 1 form a basic domain in the molecule and therefore form part of a heparin-binding site. Peptides homologous to this region of FGF receptor 1 were shown to inhibit mitogenesis in 2.3D cells, while those to FGF receptor types 2, 3, and 4 did not. A reverse transcriptase-polymerase chain reaction assay designed to detect expression of the four FGF receptors types demonstrated that FGF receptors 1 and 3 were present on the 2.3D cell line but that receptors 2 and 4 were not. These findings indicate that unique heparan sulfate domains interact with specific cell-surface receptors to direct cellular responses.
Collapse
Affiliation(s)
- Y G Brickman
- Department of Anatomy and Cell Biology, University of Melbourne, Australia
| | | | | | | | | |
Collapse
|
38
|
Seo M, Noguchi K. Retinoic acid induces gene expression of fibroblast growth factor-9 during induction of neuronal differentiation of mouse embryonal carcinoma P19 cells. FEBS Lett 1995; 370:231-5. [PMID: 7656983 DOI: 10.1016/0014-5793(95)00836-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
We have found that the gene expression of the ninth member of the fibroblast growth factor (FGF) family, FGF9 was induced during retinoic acid(RA)-induced neuronal differentiation of murine embryonal carcinoma P19 cells. We have reported here the nucleotide sequence of the mouse FGF9 cDNA. The murine cDNA showed 92.4% nucleotide sequence homology to the human FGF9 cDNA and 98.2% homology to that of rats. This mouse FGF9 cDNA encoded a polypeptide consisting of 208 amino acids with amino acid sequence identical to that of rats. Only one amino acid was replaced compared to the human homolog. The highly conserved sequence homology of FGF9 suggests its functional importance. FGF9 was originally isolated from a culture medium of a human glioma cell line as a growth-promoting factor for glial cells [5]. Upon induction of neuronal differentiation by forming cell aggregates with 10(-6) M RA, the gene expression of FGF9 was increased biphasically during the first 96 hours when cells were aggregating and from 168 hours to 192 hours followed by plating onto a tissue culture dish as glia-like cells proliferated. Neither undifferentiated P19 cells nor the cells aggregated without RA remaining undifferentiated expressed FGF9. This indicates that RA regulates the gene expression of FGF9 that may play an important role in neuronal differentiation in both early and late developmental process.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Base Sequence
- Carcinoma, Embryonal/drug therapy
- Carcinoma, Embryonal/genetics
- Carcinoma, Embryonal/pathology
- Cattle
- Cell Differentiation/drug effects
- Cell Differentiation/genetics
- DNA, Complementary/chemistry
- Fibroblast Growth Factors/drug effects
- Fibroblast Growth Factors/genetics
- Gene Expression Regulation, Developmental/drug effects
- Gene Expression Regulation, Developmental/genetics
- Humans
- Mice
- Molecular Sequence Data
- Neurons/cytology
- RNA, Messenger/drug effects
- RNA, Messenger/genetics
- Rats
- Sequence Analysis, DNA
- Tretinoin/pharmacology
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- M Seo
- Department of Biotechnology, Faculty of Engineering, Kyoto Sangyo University, Japan
| | | |
Collapse
|
39
|
Gonatas JO, Mourelatos Z, Stieber A, Lane WS, Brosius J, Gonatas NK. MG-160, a membrane sialoglycoprotein of the medial cisternae of the rat Golgi apparatus, binds basic fibroblast growth factor and exhibits a high level of sequence identity to a chicken fibroblast growth factor receptor. J Cell Sci 1995; 108 ( Pt 2):457-67. [PMID: 7768993 DOI: 10.1242/jcs.108.2.457] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We report the primary structure of MG-160, a 160 kDa membrane sialoglycoprotein residing in the medial cisternae of the Golgi apparatus of rat neurons, pheochromocytoma (PC-12), and several other cells. The cDNA encodes a polypeptide of 1,171 amino acids with an M(r) of 133,403. An intralumenal cleavable signal peptide is followed by a Pro-Gln-rich segment and 16 contiguous, approx. 60-residue-long, regularly spaced cysteine-rich segments showing sequence identities ranging from 15 to 35%. The lumenal domain is followed by a single membrane spanning domain and a short carboxy-terminal cytoplasmic tail. The protein contains 5 potential NXT glycosylation sites. The sequence of MG-160 shows no homologies with enzymes and other membrane proteins of the Golgi apparatus. MG-160 displays a so far unique feature for a membrane protein of the Golgi apparatus: namely, an upstream, open reading frame (uORF), encoding 58 amino acids, located in front of the major open reading frame (ORF). Most vertebrate mRNAs containing uORF or AUG codons in front of the major ORF encode growth factors and cell surface receptors (Geballe and Morris 1994). In that regard a 90% identity between the primary structure of MG-160 and a receptor for acidic and basic fibroblast growth factors (CFR), isolated from chicken embryos (Burrus et. al., 1992), may be relevant. Immunoreactivity for MG-160 has been detected in the Golgi apparatus of neural and other cells of 2-day-old chicken embryos and adult chicken; furthermore, recombinant human basic fibroblast growth factor (bFGF) binds MG-160 purified from rat brain. MG-160 shows no sequence similarity with members of the family of fibroblast growth factor receptors (FGFR) involved in signal transduction. These findings are consistent with the hypothesis that MG-160 is involved in the traffic and processing of endogenous or autocrine FGFs. This is the first example of an intrinsic membrane protein of the Golgi apparatus which binds a growth factor and may be involved in its regulation.
Collapse
Affiliation(s)
- J O Gonatas
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia 19104, USA
| | | | | | | | | | | |
Collapse
|
40
|
Wright DE, Snider WD. Neurotrophin receptor mRNA expression defines distinct populations of neurons in rat dorsal root ganglia. J Comp Neurol 1995; 351:329-38. [PMID: 7706545 DOI: 10.1002/cne.903510302] [Citation(s) in RCA: 238] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The biological actions of neurotrophins are mediated by specific neurotrophin receptor tyrosine kinases (Trks). A low-affinity nerve growth factor (NGF) receptor, p75, appears to modulate sensitivity to neurotrophins in some neuronal populations. It has been recently demonstrated that genes encoding members of the Trk family are expressed in distinct patterns in the dorsal root ganglia (DRG; Mu et al. [1993] (J. Neurosci. 13:4029- 4041). However, the extent to which different neurotrophin receptor genes are coexpressed by individual DRG neurons is unknown. The question of coexpression is important since the expression of more than one member of the trk family by DRG neurons would suggest the potential for regulation by multiple neurotrophins. To address this question, a combination of isotopic and colorimetric in situ hybridization was performed on rat thoracic DRG using riboprobes specific for trkA, trkB, trkC, and p75. We show here that neurons that express trkA are largely distinct from those that express trkC, although there is a small subpopulation that expresses both of these genes. We also show that there is a distinct population of DRG neurons that expresses trkB and does not coexpress either trkA or trkC. P75 is expressed in almost all neurons that express trkA or trkB, but is coexpressed in only 50% of trkC-expressing neurons. Importantly, p75 is not expressed in DRG neurons independent of trk expression. Finally, a subpopulation of DRG neurons does not express any of the neurotrophin receptor mRNAs. Our results demonstrate that there are distinct populations of DRG neurons that express each member of the neurotrophin receptor tyrosine kinase family. Our findings of extensive colocalization of p75 with trkA and trkB lend support to the idea that p75 is important in mediating the actions of NGF and brain-derived neurotrophic factor on DRG neurons. Interestingly, however, p75 expression is clearly unimportant for a subpopulation of neurons that require neurotrophin-3. The fact that p75 is not expressed in the absence of trkA, trkB, or trkC suggests that the function of p75 is closely related to functions of the known neurotrophin-receptor tyrosine kinases. Finally, our results suggest that a significant percentage of DRG neurons may be regulated by non-neurotrophin neuronal growth factors.
Collapse
Affiliation(s)
- D E Wright
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri 63110
| | | |
Collapse
|