1
|
Gasser E, Sancar G, Downes M, Evans RM. Metabolic Messengers: fibroblast growth factor 1. Nat Metab 2022; 4:663-671. [PMID: 35681108 PMCID: PMC9624216 DOI: 10.1038/s42255-022-00580-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/15/2022] [Accepted: 04/27/2022] [Indexed: 11/09/2022]
Abstract
While fibroblast growth factor (FGF) 1 is expressed in multiple tissues, only adipose-derived and brain FGF1 have been implicated in the regulation of metabolism. Adipose FGF1 production is upregulated in response to dietary stress and is essential for adipose tissue plasticity in these conditions. Similarly, in the brain, FGF1 secretion into the ventricular space and the adjacent parenchyma is increased after a hypercaloric challenge induced by either feeding or glucose infusion. Potent anorexigenic properties have been ascribed to both peripheral and centrally injected FGF1. The ability of recombinant FGF1 and variants with reduced mitogenicity to lower glucose, suppress adipose lipolysis and promote insulin sensitization elevates their potential as candidates in the treatment of type 2 diabetes mellitus and associated comorbidities. Here, we provide an overview of the known metabolic functions of endogenous FGF1 and discuss its therapeutic potential, distinguishing between peripherally or centrally administered FGF1.
Collapse
Affiliation(s)
- Emanuel Gasser
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Gencer Sancar
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Michael Downes
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Ronald M Evans
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA.
| |
Collapse
|
2
|
Ursem SR, Diepenbroek C, Bacic V, Unmehopa UA, Eggels L, Maya‐Monteiro CM, Heijboer AC, la Fleur SE. Localization of fibroblast growth factor 23 protein in the rat hypothalamus. Eur J Neurosci 2021; 54:5261-5271. [PMID: 34184338 PMCID: PMC8456796 DOI: 10.1111/ejn.15375] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 06/16/2021] [Accepted: 06/21/2021] [Indexed: 11/29/2022]
Abstract
Fibroblast growth factor 23 (FGF23) is an endocrine growth factor and known to play a pivotal role in phosphate homeostasis. Interestingly, several studies point towards a function of FGF23 in the hypothalamus. FGF23 classically activates the FGF receptor 1 in the presence of the co-receptor αKlotho, of both gene expression in the brain was previously established. However, studies on gene and protein expression of FGF23 in the brain are scarce and have been inconsistent. Therefore, our aim was to localise FGF23 gene and protein expression in the rat brain with focus on the hypothalamus. Also, we investigated the protein expression of αKlotho. Adult rat brains were used to localise and visualise FGF23 and αKlotho protein in the hypothalamus by immunofluorescence labelling. Furthermore, western blots were used for assessing hypothalamic FGF23 protein expression. FGF23 gene expression was investigated by qPCR in punches of the arcuate nucleus, lateral hypothalamus, paraventricular nucleus, choroid plexus, ventrolateral thalamic nucleus and the ventromedial hypothalamus. Immunoreactivity for FGF23 and αKlotho protein was found in the hypothalamus, third ventricle lining and the choroid plexus. Western blot analysis of the hypothalamus confirmed the presence of FGF23. Gene expression of FGF23 was not detected, suggesting that the observed FGF23 protein is not brain-derived. Several FGF receptors are known to be present in the brain. Therefore, we conclude that the machinery for FGF23 signal transduction is present in several brain areas, indeed suggesting a role for FGF23 in the brain.
Collapse
Affiliation(s)
- Stan R. Ursem
- Endocrine Laboratory, Department of Clinical Chemistry, Amsterdam Gastroenterology & MetabolismAmsterdam UMC, Vrije Universiteit Amsterdam and University of AmsterdamAmsterdamThe Netherlands
| | - Charlene Diepenbroek
- Department of Endocrinology and Metabolism and Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam NeuroscienceAmsterdam UMC, University of AmsterdamAmsterdamThe Netherlands
- Metabolism and Reward Group, Netherlands Institute for NeuroscienceAn Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW)AmsterdamThe Netherlands
| | - Vesna Bacic
- Department of Endocrinology and Metabolism and Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam NeuroscienceAmsterdam UMC, University of AmsterdamAmsterdamThe Netherlands
- Metabolism and Reward Group, Netherlands Institute for NeuroscienceAn Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW)AmsterdamThe Netherlands
| | - Unga A. Unmehopa
- Department of Endocrinology and Metabolism and Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam NeuroscienceAmsterdam UMC, University of AmsterdamAmsterdamThe Netherlands
- Metabolism and Reward Group, Netherlands Institute for NeuroscienceAn Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW)AmsterdamThe Netherlands
| | - Leslie Eggels
- Department of Endocrinology and Metabolism and Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam NeuroscienceAmsterdam UMC, University of AmsterdamAmsterdamThe Netherlands
- Metabolism and Reward Group, Netherlands Institute for NeuroscienceAn Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW)AmsterdamThe Netherlands
| | - Clarissa M. Maya‐Monteiro
- Department of Endocrinology and Metabolism and Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam NeuroscienceAmsterdam UMC, University of AmsterdamAmsterdamThe Netherlands
- Metabolism and Reward Group, Netherlands Institute for NeuroscienceAn Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW)AmsterdamThe Netherlands
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC)Oswaldo Cruz Foundation (FIOCRUZ)Rio de JaneiroBrazil
| | - Annemieke C. Heijboer
- Endocrine Laboratory, Department of Clinical Chemistry, Amsterdam Gastroenterology & MetabolismAmsterdam UMC, Vrije Universiteit Amsterdam and University of AmsterdamAmsterdamThe Netherlands
| | - Susanne E. la Fleur
- Department of Endocrinology and Metabolism and Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam NeuroscienceAmsterdam UMC, University of AmsterdamAmsterdamThe Netherlands
- Metabolism and Reward Group, Netherlands Institute for NeuroscienceAn Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW)AmsterdamThe Netherlands
| |
Collapse
|
3
|
Wean JB, Smith BN. FGF19 in the Hindbrain Lowers Blood Glucose and Alters Excitability of Vagal Motor Neurons in Hyperglycemic Mice. Endocrinology 2021; 162:6127285. [PMID: 33534906 PMCID: PMC7906449 DOI: 10.1210/endocr/bqab021] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Indexed: 12/11/2022]
Abstract
Fibroblast growth factor 19 (FGF19) is a protein hormone that produces antidiabetic effects when administered intracerebroventricularly in the forebrain. However, no studies have examined how FGF19 affects hindbrain neurons that participate directly in autonomic control of systemic glucose regulation. Within the dorsal hindbrain, parasympathetic motor neurons of the dorsal motor nucleus of the vagus (DMV) express fibroblast growth factor receptors and their activity regulates visceral homeostatic processes, including energy balance. This study tested the hypothesis that FGF19 acts in the hindbrain to alter DMV neuron excitability and lower blood glucose concentration. Fourth ventricle administration of FGF19 produced no effect on blood glucose concentration in control mice, but induced a significant, peripheral muscarinic receptor-dependent decrease in systemic hyperglycemia for up to 12 h in streptozotocin-treated mice, a model of type 1 diabetes. Patch-clamp recordings from DMV neurons in vitro revealed that FGF19 application altered synaptic and intrinsic membrane properties of DMV neurons, with the balance of FGF19 effects being significantly modified by a recent history of systemic hyperglycemia. These findings identify central parasympathetic circuitry as a novel target for FGF19 and suggest that FGF19 acting in the dorsal hindbrain can alter vagal output to produce its beneficial metabolic effects.
Collapse
Affiliation(s)
- Jordan B Wean
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Bret N Smith
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, USA
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, USA
- Correspondence: Bret N Smith, PhD, Department of Neuroscience, 800 Rose Street, Lexington, KY 40536-0298.
| |
Collapse
|
4
|
Cheng AH, Cheng HYM. Genesis of the Master Circadian Pacemaker in Mice. Front Neurosci 2021; 15:659974. [PMID: 33833665 PMCID: PMC8021851 DOI: 10.3389/fnins.2021.659974] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 03/04/2021] [Indexed: 12/13/2022] Open
Abstract
The suprachiasmatic nucleus (SCN) of the hypothalamus is the central circadian clock of mammals. It is responsible for communicating temporal information to peripheral oscillators via humoral and endocrine signaling, ultimately controlling overt rhythms such as sleep-wake cycles, body temperature, and locomotor activity. Given the heterogeneity and complexity of the SCN, its genesis is tightly regulated by countless intrinsic and extrinsic factors. Here, we provide a brief overview of the development of the SCN, with special emphasis on the murine system.
Collapse
Affiliation(s)
- Arthur H. Cheng
- Department of Biology, University of Toronto Mississauga, Mississauga, ON, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Hai-Ying Mary Cheng
- Department of Biology, University of Toronto Mississauga, Mississauga, ON, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
5
|
Wean JB, Smith BN. Fibroblast Growth Factor 19 Increases the Excitability of Pre-Motor Glutamatergic Dorsal Vagal Complex Neurons From Hyperglycemic Mice. Front Endocrinol (Lausanne) 2021; 12:765359. [PMID: 34858337 PMCID: PMC8632226 DOI: 10.3389/fendo.2021.765359] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 10/26/2021] [Indexed: 01/14/2023] Open
Abstract
Intracerebroventricular administration of the protein hormone fibroblast growth factor 19 (FGF19) to the hindbrain produces potent antidiabetic effects in hyperglycemic mice that are likely mediated through a vagal parasympathetic mechanism. FGF19 increases the synaptic excitability of parasympathetic motor neurons in the dorsal motor nucleus of the vagus (DMV) from hyperglycemic, but not normoglycemic, mice but the source of this synaptic input is unknown. Neurons in the area postrema (AP) and nucleus tractus solitarius (NTS) express high levels of FGF receptors and exert glutamatergic control over the DMV. This study tested the hypothesis that FGF19 increases glutamate release in the DMV by increasing the activity of glutamatergic AP and NTS neurons in hyperglycemic mice. Glutamate photoactivation experiments confirmed that FGF19 increases synaptic glutamate release from AP and NTS neurons that connect to the DMV in hyperglycemic, but not normoglycemic mice. Contrary to expectations, FGF19 produced a mixed effect on intrinsic membrane properties in the NTS with a trend towards inhibition, suggesting that another mechanism was responsible for the observed effects on glutamate release in the DMV. Consistent with the hypothesis, FGF19 increased action potential-dependent glutamate release in the NTS in hyperglycemic mice only. Finally, glutamate photoactivation experiments confirmed that FGF19 increases the activity of glutamatergic AP neurons that project to the NTS in hyperglycemic mice. Together, these results support the hypothesis that FGF19 increases glutamate release from AP and NTS neurons that project to the DMV in hyperglycemic mice. FGF19 therefore modifies the local vago-vagal reflex circuitry at several points. Additionally, since the AP and NTS communicate with several other metabolic regulatory nuclei in the brain, FGF19 in the hindbrain may alter neuroendocrine and behavioral aspects of metabolism, in addition to changes in parasympathetic output.
Collapse
Affiliation(s)
- Jordan B. Wean
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Bret N. Smith
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, United States
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, United States
- *Correspondence: Bret N. Smith,
| |
Collapse
|
6
|
An Overview of Nicotinic Cholinergic System Signaling in Neurogenesis. Arch Med Res 2020; 51:287-296. [DOI: 10.1016/j.arcmed.2020.03.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 03/13/2020] [Accepted: 03/30/2020] [Indexed: 12/19/2022]
|
7
|
Kumar R, Tang Q, Müller SA, Gao P, Mahlstedt D, Zampagni S, Tan Y, Klingl A, Bötzel K, Lichtenthaler SF, Höglinger GU, Koeglsperger T. Fibroblast Growth Factor 2-Mediated Regulation of Neuronal Exosome Release Depends on VAMP3/Cellubrevin in Hippocampal Neurons. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1902372. [PMID: 32195080 PMCID: PMC7080514 DOI: 10.1002/advs.201902372] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 12/11/2019] [Indexed: 05/06/2023]
Abstract
Extracellular vesicles (EVs) are endogenous membrane-derived vesicles that shuttle bioactive molecules between glia and neurons, thereby promoting neuronal survival and plasticity in the central nervous system (CNS) and contributing to neurodegenerative conditions. Although EVs hold great potential as CNS theranostic nanocarriers, the specific molecular factors that regulate neuronal EV uptake and release are currently unknown. A combination of patch-clamp electrophysiology and pH-sensitive dye imaging is used to examine stimulus-evoked EV release in individual neurons in real time. Whereas spontaneous electrical activity and the application of a high-frequency stimulus induce a slow and prolonged fusion of multivesicular bodies (MVBs) with the plasma membrane (PM) in a subset of cells, the neurotrophic factor basic fibroblast growth factor (bFGF) greatly increases the rate of stimulus-evoked MVB-PM fusion events and, consequently, the abundance of EVs in the culture medium. Proteomic analysis of neuronal EVs demonstrates bFGF increases the abundance of the v-SNARE vesicle-associated membrane protein 3 (VAMP3, cellubrevin) on EVs. Conversely, knocking-down VAMP3 in cultured neurons attenuates the effect of bFGF on EV release. The results determine the temporal characteristics of MVB-PM fusion in hippocampal neurons and reveal a new function for bFGF signaling in controlling neuronal EV release.
Collapse
Affiliation(s)
- Rohit Kumar
- Department of Translational NeurodegenerationGerman Centre for Neurodegenerative DiseasesFeodor‐Lynen‐Str. 1781377MunichGermany
- Department of NeurologyLudwig Maximilian UniversityMarchioninistr. 1581377MunichGermany
- Graduate Program for Experimental MedicineFaculty of MedicineTechnical University of MunichIsmaninger Straße 2281675MünchenGermany
| | - Qilin Tang
- Department of Translational NeurodegenerationGerman Centre for Neurodegenerative DiseasesFeodor‐Lynen‐Str. 1781377MunichGermany
- Department of NeurologyLudwig Maximilian UniversityMarchioninistr. 1581377MunichGermany
| | - Stephan A. Müller
- Department of NeuroproteomicsGerman Centre for Neurodegenerative DiseasesFeodor‐Lynen‐Str. 1781377MunichGermany
| | - Pan Gao
- Department of Translational NeurodegenerationGerman Centre for Neurodegenerative DiseasesFeodor‐Lynen‐Str. 1781377MunichGermany
| | - Diana Mahlstedt
- Department of Translational NeurodegenerationGerman Centre for Neurodegenerative DiseasesFeodor‐Lynen‐Str. 1781377MunichGermany
- Graduate Program for Experimental MedicineFaculty of MedicineTechnical University of MunichIsmaninger Straße 2281675MünchenGermany
| | - Sofia Zampagni
- Department of Translational NeurodegenerationGerman Centre for Neurodegenerative DiseasesFeodor‐Lynen‐Str. 1781377MunichGermany
| | - Yi Tan
- Department of Translational NeurodegenerationGerman Centre for Neurodegenerative DiseasesFeodor‐Lynen‐Str. 1781377MunichGermany
- Graduate Program for Experimental MedicineFaculty of MedicineTechnical University of MunichIsmaninger Straße 2281675MünchenGermany
| | - Andreas Klingl
- Plant Development and Electron MicroscopyDepartment of Biology IBiocenterLudwig Maximilian UniversityGroßhaderner Str. 282152Planegg‐MartinsriedGermany
| | - Kai Bötzel
- Department of NeurologyLudwig Maximilian UniversityMarchioninistr. 1581377MunichGermany
| | - Stefan F. Lichtenthaler
- Department of NeuroproteomicsGerman Centre for Neurodegenerative DiseasesFeodor‐Lynen‐Str. 1781377MunichGermany
- NeuroproteomicsKlinikum rechts der IsarInstitute for Advanced StudyTechnical University of MunichIsmaninger Straße 2281675MunichGermany
| | - Günter U. Höglinger
- Department of Translational NeurodegenerationGerman Centre for Neurodegenerative DiseasesFeodor‐Lynen‐Str. 1781377MunichGermany
- Department of Neurology (OE 7210)Hannover Medical SchoolCarl‐Neuberg‐Str. 130625HannoverGermany
- Department of NeurologyTechnical University of MunichIsmaninger Str. 2281675MunichGermany
| | - Thomas Koeglsperger
- Department of Translational NeurodegenerationGerman Centre for Neurodegenerative DiseasesFeodor‐Lynen‐Str. 1781377MunichGermany
- Department of NeurologyLudwig Maximilian UniversityMarchioninistr. 1581377MunichGermany
| |
Collapse
|
8
|
Kano M, Suga H, Ishihara T, Sakakibara M, Soen M, Yamada T, Ozaki H, Mitsumoto K, Kasai T, Sugiyama M, Onoue T, Tsunekawa T, Takagi H, Hagiwara D, Ito Y, Iwama S, Goto M, Banno R, Arima H. Tanycyte-Like Cells Derived From Mouse Embryonic Stem Culture Show Hypothalamic Neural Stem/Progenitor Cell Functions. Endocrinology 2019; 160:1701-1718. [PMID: 31135891 DOI: 10.1210/en.2019-00105] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 05/22/2019] [Indexed: 01/01/2023]
Abstract
Tanycytes have recently been accepted as neural stem/progenitor cells in the postnatal hypothalamus. Persistent retina and anterior neural fold homeobox (Rax) expression is characteristic of tanycytes in contrast to its transient expression of whole hypothalamic precursors. In this study, we found that Rax+ residual cells in the maturation phase of hypothalamic differentiation in mouse embryonic stem cell (mESC) cultures had similar characteristics to ventral tanycytes. They expressed typical neural stem/progenitor cell markers, including Sox2, vimentin, and nestin, and differentiated into mature neurons and glial cells. Quantitative RT-PCR analysis showed that Rax+ residual cells expressed Fgf-10, Fgf-18, and Lhx2, which are expressed by ventral tanycytes. They highly expressed tanycyte-specific genes Dio2 and Gpr50 compared with Rax+ early hypothalamic progenitor cells. Therefore, Rax+ residual cells in the maturation phase of hypothalamic differentiation were considered to be more differentiated and similar to late progenitor cells and tanycytes. They self-renewed and formed neurospheres when cultured with exogenous FGF-2. Additionally, these Rax+ neurospheres differentiated into three neuronal lineages (neurons, astrocytes, and oligodendrocytes), including neuropeptide Y+ neuron, that are reported to be differentiated from ventral tanycytes toward the arcuate nuclei. Thus, Rax+ residual cells were multipotent neural stem/progenitor cells. Rax+ neurospheres were stably passaged and retained high Sox2 expression even after multiple passages. These results suggest the successful induction of Rax+ tanycyte-like cells from mESCs [induced tanycyte-like (iTan) cells]. These hypothalamic neural stem/progenitor cells may have potential in regenerative medicine and as a research tool.
Collapse
Affiliation(s)
- Mayuko Kano
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hidetaka Suga
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takeshi Ishihara
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Drug Discovery Technologies, Drug Discovery and Disease Research Laboratory, Shionogi and Co., Ltd., Osaka, Japan
| | - Mayu Sakakibara
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mika Soen
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomiko Yamada
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hajime Ozaki
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazuki Mitsumoto
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takatoshi Kasai
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mariko Sugiyama
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takeshi Onoue
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Taku Tsunekawa
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroshi Takagi
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Daisuke Hagiwara
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshihiro Ito
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shintaro Iwama
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Motomitsu Goto
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ryoichi Banno
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroshi Arima
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
9
|
Cross-Talk between Fibroblast Growth Factor Receptors and Other Cell Surface Proteins. Cells 2019; 8:cells8050455. [PMID: 31091809 PMCID: PMC6562592 DOI: 10.3390/cells8050455] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 05/08/2019] [Accepted: 05/13/2019] [Indexed: 12/14/2022] Open
Abstract
Fibroblast growth factors (FGFs) and their receptors (FGFRs) constitute signaling circuits that transmit signals across the plasma membrane, regulating pivotal cellular processes like differentiation, migration, proliferation, and apoptosis. The malfunction of FGFs/FGFRs signaling axis is observed in numerous developmental and metabolic disorders, and in various tumors. The large diversity of FGFs/FGFRs functions is attributed to a great complexity in the regulation of FGFs/FGFRs-dependent signaling cascades. The function of FGFRs is modulated at several levels, including gene expression, alternative splicing, posttranslational modifications, and protein trafficking. One of the emerging ways to adjust FGFRs activity is through formation of complexes with other integral proteins of the cell membrane. These proteins may act as coreceptors, modulating binding of FGFs to FGFRs and defining specificity of elicited cellular response. FGFRs may interact with other cell surface receptors, like G-protein-coupled receptors (GPCRs) or receptor tyrosine kinases (RTKs). The cross-talk between various receptors modulates the strength and specificity of intracellular signaling and cell fate. At the cell surface FGFRs can assemble into large complexes involving various cell adhesion molecules (CAMs). The interplay between FGFRs and CAMs affects cell–cell interaction and motility and is especially important for development of the central nervous system. This review summarizes current stage of knowledge about the regulation of FGFRs by the plasma membrane-embedded partner proteins and highlights the importance of FGFRs-containing membrane complexes in pathological conditions, including cancer.
Collapse
|
10
|
Brown JM, Scarlett JM, Matsen ME, Nguyen HT, Secher A, Jorgensen R, Morton GJ, Schwartz MW. The Hypothalamic Arcuate Nucleus-Median Eminence Is a Target for Sustained Diabetes Remission Induced by Fibroblast Growth Factor 1. Diabetes 2019; 68:1054-1061. [PMID: 30796029 PMCID: PMC6477902 DOI: 10.2337/db19-0025] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 02/14/2019] [Indexed: 12/13/2022]
Abstract
In rodent models of type 2 diabetes (T2D), sustained remission of diabetic hyperglycemia can be induced by a single intracerebroventricular (icv) injection of fibroblast growth factor 1 (FGF1). To identify the brain areas responsible for this effect, we first used immunohistochemistry to map the hypothalamic distribution of phosphorylated extracellular signal-related kinase 1/2 (pERK1/2), a marker of mitogen-activated protein kinase-ERK signal transduction downstream of FGF receptor activation. Twenty minutes after icv FGF1 injection in adult male Wistar rats, pERK1/2 staining was detected primarily in two hypothalamic areas: the arcuate nucleus-median eminence (ARC-ME) and the paraventricular nucleus (PVN). To determine whether an action of FGF1 localized to either the ARC-ME or the PVN is capable of mimicking the sustained antidiabetic effect elicited by icv FGF1, we microinjected either saline vehicle or a low dose of FGF1 (0.3 µg/side) bilaterally into either the ARC-ME area or PVN of Zucker Diabetic Fatty rats, a model of T2D, and monitored daily food intake, body weight, and blood glucose levels over a 3-week period. Whereas bilateral intra-arcuate microinjection of saline vehicle was without effect, remission of hyperglycemia lasting >3 weeks was observed following bilateral microinjection of FGF1 into the ARC-ME. This antidiabetic effect cannot be attributed to leakage of FGF1 into cerebrospinal fluid and subsequent action on other brain areas, since icv injection of the same total dose was without effect. Combined with our finding that bilateral microinjection of the same dose of FGF1 into the PVN was without effect on glycemia or other parameters, we conclude that the ARC-ME area (but not the PVN) is a target for sustained remission of diabetic hyperglycemia induced by FGF1.
Collapse
Affiliation(s)
- Jenny M Brown
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle, WA
| | - Jarrad M Scarlett
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle, WA
- Department of Pediatric Gastroenterology and Hepatology, University of Washington, Seattle, WA
| | - Miles E Matsen
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle, WA
| | - Hong T Nguyen
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle, WA
| | - Anna Secher
- Diabetes Research, Global Drug Discovery, Novo Nordisk, Måløv, Denmark
| | - Rasmus Jorgensen
- Diabetes Research, Global Drug Discovery, Novo Nordisk, Måløv, Denmark
| | - Gregory J Morton
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle, WA
| | - Michael W Schwartz
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle, WA
| |
Collapse
|
11
|
Hultman K, Scarlett JM, Baquero AF, Cornea A, Zhang Y, Salinas CBG, Brown J, Morton GJ, Whalen EJ, Grove KL, Koegler FH, Schwartz MW, Mercer AJ. The central fibroblast growth factor receptor/beta klotho system: Comprehensive mapping in Mus musculus and comparisons to nonhuman primate and human samples using an automated in situ hybridization platform. J Comp Neurol 2019; 527:2069-2085. [PMID: 30809795 DOI: 10.1002/cne.24668] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 02/20/2019] [Accepted: 02/20/2019] [Indexed: 12/25/2022]
Abstract
Central activation of fibroblast growth factor (FGF) receptors regulates peripheral glucose homeostasis and reduces food intake in preclinical models of obesity and diabetes. The current work was undertaken to advance our understanding of the receptor expression, as sites of ligand action by FGF19, FGF21, and FGF1 in the mammalian brain remains unresolved. Recent advances in automated RNAscope in situ hybridization and droplet digital PCR (ddPCR) technology allowed us to interrogate central FGFR/beta klotho (Klb) system at the cellular level in the mouse, with relevant comparisons to nonhuman primate and human brain. FGFR1-3 gene expression was broadly distributed throughout the CNS in Mus musculus, with FGFR1 exhibiting the greatest heterogeneity. FGFR4 expression localized only in the medial habenula and subcommissural organ of mice. Likewise, Klb mRNA was restricted to the suprachiasmatic nucleus (SCh) and select midbrain and hindbrain nuclei. ddPCR in the rodent hypothalamus confirmed that, although expression levels are indeed low for Klb, there is nonetheless a bonafide subpopulation of Klb+ cells in the hypothalamus. In NHP and human midbrain and hindbrain, Klb + cells are quite rare, as is expression of FGFR4. Collectively, these data provide the most robust central map of the FGFR/Klb system to date and highlight central regions that may be of critical importance to assess central ligand effects with pharmacological dosing, such as the putative interactions between the endocrine FGFs and FGFR1/Klb, or FGF19 with FGFR4.
Collapse
Affiliation(s)
| | - Jarrad M Scarlett
- Diabetes & Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, Washington.,Department of Pediatric Gastroenterology & Hepatology, Seattle Children's Hospital, Seattle, Washington
| | - Arian F Baquero
- Novo Nordisk Research Center Seattle, Inc., Seattle, Washington
| | - Anda Cornea
- Novo Nordisk Research Center Seattle, Inc., Seattle, Washington
| | - Yu Zhang
- Novo Nordisk Research Center Seattle, Inc., Seattle, Washington
| | | | - Jenny Brown
- Diabetes & Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, Washington
| | - Gregory J Morton
- Diabetes & Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, Washington
| | - Erin J Whalen
- Novo Nordisk Research Center Seattle, Inc., Seattle, Washington
| | - Kevin L Grove
- Novo Nordisk Research Center Seattle, Inc., Seattle, Washington
| | - Frank H Koegler
- Novo Nordisk Research Center Seattle, Inc., Seattle, Washington
| | - Michael W Schwartz
- Diabetes & Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, Washington
| | - Aaron J Mercer
- Novo Nordisk Research Center Seattle, Inc., Seattle, Washington
| |
Collapse
|
12
|
Langlet F. Tanycyte Gene Expression Dynamics in the Regulation of Energy Homeostasis. Front Endocrinol (Lausanne) 2019; 10:286. [PMID: 31133987 PMCID: PMC6514105 DOI: 10.3389/fendo.2019.00286] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 04/18/2019] [Indexed: 01/01/2023] Open
Abstract
Animal survival relies on a constant balance between energy supply and energy expenditure, which is controlled by several neuroendocrine functions that integrate metabolic information and adapt the response of the organism to physiological demands. Polarized ependymoglial cells lining the floor of the third ventricle and sending a single process within metabolic hypothalamic parenchyma, tanycytes are henceforth described as key components of the hypothalamic neural network controlling energy balance. Their strategic position and peculiar properties convey them diverse physiological functions ranging from blood/brain traffic controllers, metabolic modulators, and neural stem/progenitor cells. At the molecular level, these functions rely on an accurate regulation of gene expression. Indeed, tanycytes are characterized by their own molecular signature which is mostly associated to their diverse physiological functions, and the detection of variations in nutrient/hormone levels leads to an adequate modulation of genetic profile in order to ensure energy homeostasis. The aim of this review is to summarize recent knowledge on the nutritional control of tanycyte gene expression.
Collapse
|
13
|
Di Liberto V, Mudò G, Belluardo N. Crosstalk between receptor tyrosine kinases (RTKs) and G protein-coupled receptors (GPCR) in the brain: Focus on heteroreceptor complexes and related functional neurotrophic effects. Neuropharmacology 2018; 152:67-77. [PMID: 30445101 DOI: 10.1016/j.neuropharm.2018.11.018] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 11/01/2018] [Accepted: 11/12/2018] [Indexed: 01/11/2023]
Abstract
Neuronal events are regulated by the integration of several complex signaling networks in which G protein-coupled receptors (GPCRs) and receptor tyrosine kinases (RTKs) are considered key players of an intense bidirectional cross-communication in the cell, generating signaling mechanisms that, at the same time, connect and diversify the traditional signal transduction pathways activated by the single receptor. For this receptor-receptor crosstalk, the two classes of receptors form heteroreceptor complexes resulting in RTKs transactivation and in growth-promoting signals. In this review, we describe heteroreceptor complexes between GPCR and RTKs in the central nervous system (CNS) and their functional effects in controlling a variety of neuronal effects, ranging from development, proliferation, differentiation and migration, to survival, repair, synaptic transmission and plasticity. In this interaction, RTKs can also recruit components of the G protein signaling cascade, creating a bidirectional intricate interplay that provides complex control over multiple cellular events. These heteroreceptor complexes, by the integration of different signals, have recently attracted a growing interest as novel molecular target for depressive disorders. This article is part of the Special Issue entitled 'Receptor heteromers and their allosteric receptor-receptor interactions'.
Collapse
Affiliation(s)
- Valentina Di Liberto
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy
| | - Giuseppa Mudò
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy
| | - Natale Belluardo
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy.
| |
Collapse
|
14
|
Even-Chen O, Barak S. The role of fibroblast growth factor 2 in drug addiction. Eur J Neurosci 2018; 50:2552-2561. [PMID: 30144335 DOI: 10.1111/ejn.14133] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 08/08/2018] [Accepted: 08/16/2018] [Indexed: 12/21/2022]
Abstract
Fibroblast growth factor 2 (FGF2) is a member of the FGF-family, which consists of 22 members, with four known FGF receptors (five in humans). Over the last 30 years, FGF2 has been extensively studied for its role in cell proliferation, differentiation, growth, survival and angiogenesis during development, as well as for its role in adult neurogenesis and regenerative plasticity. Over the past decade, FGF2 has been implicated in learning and memory, as well as in several neuropsychiatric disorders, including anxiety, stress, depression and drug addiction. In this review, we present accumulating evidence indicating the involvement of FGF2 in neuroadaptations caused by drugs of abuse, namely, amphetamine, cocaine, nicotine and alcohol. Moreover, evidence suggests that FGF2 is a positive regulator of alcohol and drug-related behaviors. Thus, although additional studies are yet required, we suggest that reducing FGF2 activity may provide a novel therapeutic approach for substance use disorders.
Collapse
Affiliation(s)
- Oren Even-Chen
- School of Psychological Sciences, Tel Aviv University, 69978, Tel Aviv, Israel
| | - Segev Barak
- School of Psychological Sciences, Tel Aviv University, 69978, Tel Aviv, Israel.,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
15
|
Methylation changes and aberrant expression of FGFR3 in Lewy body disease neurons. Brain Res 2018; 1697:59-66. [PMID: 29909202 DOI: 10.1016/j.brainres.2018.06.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 06/12/2018] [Accepted: 06/13/2018] [Indexed: 12/17/2022]
Abstract
Lewy body disease (LBD) is characterized by accumulation of aggregated α-synuclein in the central nervous system as eosinophilic cytoplasmic inclusions called Lewy bodies. According to their distribution pattern, it is classified into brainstem LBD, limbic LBD and diffuse neocortical LBD. It has been reported that α-synuclein affects various points in the MAPK cascade but its relationship with FGF receptors, which are the most upstream of the pathway, has not been previously investigated. We discovered that among the four FGFRs, FGFR3 showed neuronal upregulation in LBD brains histopathologically. Further examination using neuron-specific methylome analysis revealed that the gene body of FGFR3 was hypermethylated in LBD, suggesting its increased transcription. Altered methylation was not observed in the non-neuronal genome. Altered methylation status was associated with the severity of α-synuclein pathology.
Collapse
|
16
|
Boshoff EL, Fletcher EJR, Duty S. Fibroblast growth factor 20 is protective towards dopaminergic neurons in vivo in a paracrine manner. Neuropharmacology 2018; 137:156-163. [PMID: 29698669 PMCID: PMC6063078 DOI: 10.1016/j.neuropharm.2018.04.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 04/18/2018] [Accepted: 04/19/2018] [Indexed: 01/01/2023]
Abstract
Neuroprotective strategies are an unmet medical need for Parkinson's disease. Fibroblast growth factor 20 (FGF20) enhances survival of cultured dopaminergic neurons but little is known about its in vivo potential. We set out to examine whether manipulation of the FGF20 system affected nigrostriatal tract integrity in rats, to identify which fibroblast growth factor receptors (FGFRs) might reside on dopaminergic neurons and to discover the source of endogenous FGF20 in the substantia nigra (SN). Male Sprague Dawley rats were subject to a partial 6-OHDA lesion alongside treatment with exogenous FGF20 or an FGFR antagonist. Behavioural readouts and tyrosine-hydroxylase (TH) immunohistochemistry were used to evaluate nigrostriatal tract integrity. Fluorescent immunohistochemistry was used to examine FGFR subtype expression on TH-positive dopamine neurons and FGF20 cellular localisation within the SN. FGF20 (2.5 μg/day) significantly protected TH-positive cells in the SN and terminals in the striatum, while reducing the development of motor asymmetry at 5, 8 and 11 days post lesion. Conversely, the FGFR antagonist PD173074 (2 mg/kg) significantly worsened both the 6-OHDA lesion and resultant motor asymmetry. Within the SN, TH-positive cells expressed FGFR1, 3 and 4 while FGF20 co-localised with GFAP-positive astrocytes. In conclusion, FGF20 protects dopaminergic neurons in vivo, an action likely mediated through activation of FGFRs1, 3 or 4 found on these neurons. Given FGF20 is localised to astrocytes in the adult SN, endogenous FGF20 provides its protection of dopamine neurons through a paracrine action. Boosting the endogenous FGF20 production might offer potential as a future therapeutic strategy in Parkinson's disease. FGF20 infusion protects against a partial 6-OHDA nigrostriatal tract lesion in rats. Dopaminergic cells in the SNc express FGFR1, 3 and 4 upon which FGF20 can act. FGFR blockade exacerbates a partial 6-OHDA nigrostriatal tract lesion in rats. Endogenous FGF20 is expressed by astrocytes in the SN, not by dopaminergic neurons. Endogenous FGF20 most likely provides protection in a paracrine manner.
Collapse
Affiliation(s)
- Eugene L Boshoff
- King's College London, Institute of Psychiatry, Psychology & Neuroscience, Wolfson Centre for Age-Related Diseases, Guy's Campus, London SE1 1UL, UK
| | - Edward J R Fletcher
- King's College London, Institute of Psychiatry, Psychology & Neuroscience, Wolfson Centre for Age-Related Diseases, Guy's Campus, London SE1 1UL, UK
| | - Susan Duty
- King's College London, Institute of Psychiatry, Psychology & Neuroscience, Wolfson Centre for Age-Related Diseases, Guy's Campus, London SE1 1UL, UK.
| |
Collapse
|
17
|
Ledwon JK, Turin SY, Gosain AK, Topczewska JM. The expression of fgfr3 in the zebrafish head. Gene Expr Patterns 2018; 29:32-38. [PMID: 29630949 DOI: 10.1016/j.gep.2018.04.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 03/08/2018] [Accepted: 04/03/2018] [Indexed: 12/29/2022]
Abstract
Fibroblast growth factor (FGF) signaling is essential for many developmental processes and plays a pivotal role in skeletal homeostasis, regeneration and wound healing. FGF signals through one of five tyrosine kinase receptors: Fgfr1a, -1b, -2, -3, -4. To characterize the expression of zebrafish fgfr3 from the larval stage to adulthood, we used RNAscope in situ hybridization on paraffin sections of the zebrafish head. Our study revealed spatial and temporal distribution of fgfr3 transcript in chondrocytes of the head cartilages, osteoblasts involved in bone formation, ventricular zone of the brain, undifferentiated mesenchymal cells of the skin, and lens epithelium of the eye. In general, the expression pattern of zebrafish fgfr3 is similar to the expression observed in higher vertebrates.
Collapse
Affiliation(s)
- Joanna K Ledwon
- Northwestern University Feinberg School of Medicine, Ann & Robert H. Lurie Children's Hospital of Chicago, Division of Plastic and Reconstructive Surgery, Stanley Manne Children's Research Institute, Chicago, IL, USA
| | - Sergey Y Turin
- Northwestern University Feinberg School of Medicine, Ann & Robert H. Lurie Children's Hospital of Chicago, Division of Plastic and Reconstructive Surgery, Stanley Manne Children's Research Institute, Chicago, IL, USA
| | - Arun K Gosain
- Northwestern University Feinberg School of Medicine, Ann & Robert H. Lurie Children's Hospital of Chicago, Division of Plastic and Reconstructive Surgery, Stanley Manne Children's Research Institute, Chicago, IL, USA
| | - Jolanta M Topczewska
- Northwestern University Feinberg School of Medicine, Ann & Robert H. Lurie Children's Hospital of Chicago, Division of Plastic and Reconstructive Surgery, Stanley Manne Children's Research Institute, Chicago, IL, USA.
| |
Collapse
|
18
|
Mertens KL, Kalsbeek A, Soeters MR, Eggink HM. Bile Acid Signaling Pathways from the Enterohepatic Circulation to the Central Nervous System. Front Neurosci 2017; 11:617. [PMID: 29163019 PMCID: PMC5681992 DOI: 10.3389/fnins.2017.00617] [Citation(s) in RCA: 178] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 10/23/2017] [Indexed: 12/14/2022] Open
Abstract
Bile acids are best known as detergents involved in the digestion of lipids. In addition, new data in the last decade have shown that bile acids also function as gut hormones capable of influencing metabolic processes via receptors such as FXR (farnesoid X receptor) and TGR5 (Takeda G protein-coupled receptor 5). These effects of bile acids are not restricted to the gastrointestinal tract, but can affect different tissues throughout the organism. It is still unclear whether these effects also involve signaling of bile acids to the central nervous system (CNS). Bile acid signaling to the CNS encompasses both direct and indirect pathways. Bile acids can act directly in the brain via central FXR and TGR5 signaling. In addition, there are two indirect pathways that involve intermediate agents released upon interaction with bile acids receptors in the gut. Activation of intestinal FXR and TGR5 receptors can result in the release of fibroblast growth factor 19 (FGF19) and glucagon-like peptide 1 (GLP-1), both capable of signaling to the CNS. We conclude that when plasma bile acids levels are high all three pathways may contribute in signal transmission to the CNS. However, under normal physiological circumstances, the indirect pathway involving GLP-1 may evoke the most substantial effect in the brain.
Collapse
Affiliation(s)
- Kim L Mertens
- Master's Program in Biomedical Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Andries Kalsbeek
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Laboratory of Endocrinology, Department Clinical Chemistry, Academic Medical Centre, University of Amsterdam, Amsterdam, Netherlands.,Department of Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
| | - Maarten R Soeters
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Hannah M Eggink
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Department of Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
| |
Collapse
|
19
|
Collette JC, Choubey L, Smith KM. -Glial and stem cell expression of murine Fibroblast Growth Factor Receptor 1 in the embryonic and perinatal nervous system. PeerJ 2017; 5:e3519. [PMID: 28674667 PMCID: PMC5493973 DOI: 10.7717/peerj.3519] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 06/08/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Fibroblast growth factors (FGFs) and their receptors (FGFRs) are involved in the development and function of multiple organs and organ systems, including the central nervous system (CNS). FGF signaling via FGFR1, one of the three FGFRs expressed in the CNS, stimulates proliferation of stem cells during prenatal and postnatal neurogenesis and participates in regulating cell-type ratios in many developing regions of the brain. Anomalies in FGFR1 signaling have been implicated in certain neuropsychiatric disorders. Fgfr1 expression has been shown, via in situ hybridization, to vary spatially and temporally throughout embryonic and postnatal development of the brain. However, in situ hybridization lacks sufficient resolution to identify which cell-types directly participate in FGF signaling. Furthermore, because antibodies raised against FGFR1 commonly cross-react with other members of the FGFR family, immunocytochemistry is not alone sufficient to accurately document Fgfr1 expression. Here, we elucidate the identity of Fgfr1 expressing cells in both the embryonic and perinatal mouse brain. METHODS To do this, we utilized a tgFGFR1-EGFPGP338Gsat BAC line (tgFgfr1-EGFP+) obtained from the GENSAT project. The tgFgfr1-EGFP+ line expresses EGFP under the control of a Fgfr1 promoter, thereby causing cells endogenously expressing Fgfr1 to also present a positive GFP signal. Through simple immunostaining using GFP antibodies and cell-type specific antibodies, we were able to accurately determine the cell-type of Fgfr1 expressing cells. RESULTS This technique revealed Fgfr1 expression in proliferative zones containing BLBP+ radial glial stem cells, such as the cortical and hippocampal ventricular zones, and cerebellar anlage of E14.5 mice, in addition to DCX+ neuroblasts. Furthermore, our data reveal Fgfr1 expression in proliferative zones containing BLBP+ cells of the anterior midline, hippocampus, cortex, hypothalamus, and cerebellum of P0.5 mice, in addition to the early-formed GFAP+ astrocytes of the anterior midline. DISCUSSION Understanding when during development and where Fgfr1 is expressed is critical to improving our understanding of its function during neurodevelopment as well as in the mature CNS. This information may one day provide an avenue of discovery towards understanding the involvement of aberrant FGF signaling in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Jantzen C Collette
- Department of Biology, University of Louisiana at Lafayette, Lafayette, LA, United States of America
| | - Lisha Choubey
- Department of Biology, University of Louisiana at Lafayette, Lafayette, LA, United States of America
| | - Karen Müller Smith
- Department of Biology, University of Louisiana at Lafayette, Lafayette, LA, United States of America
| |
Collapse
|
20
|
Choubey L, Collette JC, Smith KM. Quantitative assessment of fibroblast growth factor receptor 1 expression in neurons and glia. PeerJ 2017; 5:e3173. [PMID: 28439461 PMCID: PMC5398288 DOI: 10.7717/peerj.3173] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 03/13/2017] [Indexed: 01/23/2023] Open
Abstract
Background Fibroblast growth factors (FGFs) and their receptors (FGFRs) have numerous functions in the developing and adult central nervous system (CNS). For example, the FGFR1 receptor is important for proliferation and fate specification of radial glial cells in the cortex and hippocampus, oligodendrocyte proliferation and regeneration, midline glia morphology and soma translocation, Bergmann glia morphology, and cerebellar morphogenesis. In addition, FGFR1 signaling in astrocytes is required for postnatal maturation of interneurons expressing parvalbumin (PV). FGFR1 is implicated in synapse formation in the hippocampus, and alterations in the expression of Fgfr1 and its ligand, Fgf2 accompany major depression. Understanding which cell types express Fgfr1 during development may elucidate its roles in normal development of the brain as well as illuminate possible causes of certain neuropsychiatric disorders. Methods Here, we used a BAC transgenic reporter line to trace Fgfr1 expression in the developing postnatal murine CNS. The specific transgenic line employed was created by the GENSAT project, tgFGFR1-EGFPGP338Gsat, and includes a gene encoding enhanced green fluorescent protein (EGFP) under the regulation of the Fgfr1 promoter, to trace Fgfr1 expression in the developing CNS. Unbiased stereological counts were performed for several cell types in the cortex and hippocampus. Results This model reveals that Fgfr1 is primarily expressed in glial cells, in both astrocytes and oligodendrocytes, along with some neurons. Dual labeling experiments indicate that the proportion of GFP+ (Fgfr1+) cells that are also GFAP+ increases from postnatal day 7 (P7) to 1 month, illuminating dynamic changes in Fgfr1 expression during postnatal development of the cortex. In postnatal neurogenic areas, GFP expression was also observed in SOX2, doublecortin (DCX), and brain lipid-binding protein (BLBP) expressing cells. Fgfr1 is also highly expressed in DCX positive cells of the dentate gyrus (DG), but not in the rostral migratory stream. Fgfr1 driven GFP was also observed in tanycytes and GFAP+ cells of the hypothalamus, as well as in Bergmann glia and astrocytes of the cerebellum. Conclusions The tgFGFR1-EGFPGP338Gsat mouse model expresses GFP that is congruent with known functions of FGFR1, including hippocampal development, glial cell development, and stem cell proliferation. Understanding which cell types express Fgfr1 may elucidate its role in neuropsychiatric disorders and brain development.
Collapse
Affiliation(s)
- Lisha Choubey
- Department of Biology, University of Louisiana at Lafayette, United States of America
| | - Jantzen C Collette
- Department of Biology, University of Louisiana at Lafayette, United States of America
| | - Karen Müller Smith
- Department of Biology, University of Louisiana at Lafayette, United States of America
| |
Collapse
|
21
|
Di Liberto V, Borroto-Escuela DO, Frinchi M, Verdi V, Fuxe K, Belluardo N, Mudò G. Existence of muscarinic acetylcholine receptor (mAChR) and fibroblast growth factor receptor (FGFR) heteroreceptor complexes and their enhancement of neurite outgrowth in neural hippocampal cultures. Biochim Biophys Acta Gen Subj 2016; 1861:235-245. [PMID: 27815219 DOI: 10.1016/j.bbagen.2016.10.026] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 10/15/2016] [Accepted: 10/31/2016] [Indexed: 12/19/2022]
Abstract
BACKGROUND Recently, it was demonstrated that G-protein-coupled receptors (GPCRs) can transactivate tyrosine kinase receptors in absence of their ligands. In this work, driven by the observation that mAChRs and fibroblast growth factor receptors (FGFRs) share signalling pathways and regulation of brain functions, it was decided to explore whether mAChRs activation may transactivate FGFRs and, if so, to characterize the related trophic effects in cultured hippocampal neurons. METHODS Oxotremorine-M transactivation of FGFRs and related trophic effects were tested in primary hippocampal neurons. Western blotting and in situ proximity ligation assay (PLA) were used to detect FGFR phosphorylation (pFGFR) levels and M1R-FGFR1 heteroreceptor complexes, respectively. RESULTS Oxotremorine-M, a non-selective mAChRs agonist, was able to transactivate FGFR and this transactivation was blocked by Src inhibitors. Oxotremorine-M treatment produced a significant increase in the primary neurite outgrowth that was blocked by pre-treatment with the pFGFR inhibitor SU5402 and Src inhibitors. This trophic effect was almost similar to that induced by fibroblast growth factor-2 (FGF-2). By using atropine as nonselective mAChRs or pirenzepine as selective antagonist for M1 receptor (M1R) we could show that mAChRs are involved in modulating the pFGFRs. Using PLA, M1R-FGFR1 heteroreceptor complexes were identified in the hippocampus and cerebral cortex. CONCLUSION The current findings, by showing functional mAChR-FGFR interactions, will contribute to advance the understanding of the mechanisms involved in the actions of cholinergic drugs on neuronal plasticity. GENERAL SIGNIFICANT Data may help to develop novel therapeutic strategies not only for neurodegenerative diseases but also for depression-induced atrophy of hippocampal neurons.
Collapse
Affiliation(s)
- V Di Liberto
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Corso Tukory 129, 90134 Palermo, Italy.
| | - D O Borroto-Escuela
- Karolinska Instituet, Department of Neuroscience, Retzius väg 8, 17177 Stockholm, Sweden; Department of Biomolecular Science, Section of Physiology, University of Urbino, Campus Scientifico Enrico Mattei, via Ca' le Suore 2, I-61029 Urbino, Italy; Observatorio Cubano de Neurociencias, Grupo Bohío-Estudio, Zayas 50, 62100 Yaguajay, Cuba.
| | - M Frinchi
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Corso Tukory 129, 90134 Palermo, Italy.
| | - V Verdi
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Corso Tukory 129, 90134 Palermo, Italy.
| | - K Fuxe
- Karolinska Instituet, Department of Neuroscience, Retzius väg 8, 17177 Stockholm, Sweden.
| | - N Belluardo
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Corso Tukory 129, 90134 Palermo, Italy.
| | - G Mudò
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Corso Tukory 129, 90134 Palermo, Italy.
| |
Collapse
|
22
|
Salmaso N, Stevens HE, McNeill J, ElSayed M, Ren Q, Maragnoli ME, Schwartz ML, Tomasi S, Sapolsky RM, Duman R, Vaccarino FM. Fibroblast Growth Factor 2 Modulates Hypothalamic Pituitary Axis Activity and Anxiety Behavior Through Glucocorticoid Receptors. Biol Psychiatry 2016; 80:479-489. [PMID: 27133954 PMCID: PMC8009045 DOI: 10.1016/j.biopsych.2016.02.026] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 02/15/2016] [Accepted: 02/16/2016] [Indexed: 01/07/2023]
Abstract
BACKGROUND Despite strong evidence linking fibroblast growth factor 2 (FGF2) with anxiety and depression in both rodents and humans, the molecular mechanisms linking FGF2 with anxiety are not understood. METHODS We compare 1) mice that lack a functional Fgf2 gene (Fgf2 knockout [KO]), 2) wild-type mice, and 3) Fgf2 KO with adult rescue by FGF2 administration on measures of anxiety, depression, and motor behavior, and further investigate the mechanisms of this behavior by cellular, molecular, and neuroendocrine studies. RESULTS We demonstrate that Fgf2 KO mice have increased anxiety, decreased hippocampal glucocorticoid receptor (GR) expression, and increased hypothalamic-pituitary-adrenal axis activity. FGF2 administration in adulthood was sufficient to rescue the entire phenotype. Blockade of GR in adult mice treated with FGF2 precluded the therapeutic effects of FGF2 on anxiety behavior, suggesting that GR is necessary for FGF2 to regulate anxiety behavior. The level of Egr-1/NGFI-A was decreased in Fgf2 KO mice and was reestablished with FGF2 treatment. By chromatin immunoprecipitation studies, we found decreased binding of EGR-1 to the GR promoter region in Fgf2 KO mice. Finally, we examined anxiety behavior in FGF receptor (FGFR) KO mice; however, FGFR1, FGFR2, and FGFR3 KO mice did not mimic the phenotype of Fgf2 KO mice, suggesting a role for other receptor subtypes (i.e., FGFR5). CONCLUSIONS These data suggest that FGF2 levels are critically related to anxiety behavior and hypothalamic-pituitary-adrenal axis activity, likely through modulation of hippocampal glucocorticoid receptor expression, an effect that is likely receptor mediated, albeit not by FGFR1, FGFR2, and FGFR3.
Collapse
|
23
|
Borroto-Escuela DO, Tarakanov AO, Fuxe K. FGFR1-5-HT1A Heteroreceptor Complexes: Implications for Understanding and Treating Major Depression. Trends Neurosci 2015; 39:5-15. [PMID: 26687454 DOI: 10.1016/j.tins.2015.11.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Revised: 10/15/2015] [Accepted: 11/10/2015] [Indexed: 11/17/2022]
Abstract
The serotonin and neurotrophic factor hypotheses of depression are well known. The discovery of brain fibroblast growth factor receptor 1 (FGFR1)-5 hydroxytryptamine receptor 1A (5-HT1A) heteroreceptor complexes, and their enhancement of neuroplasticity, offers an integration of these hypotheses at the molecular level. They were first described in the hippocampus and later in midbrain 5-HT neurons, where these heterocomplexes are enriched in 5-HT1A autoreceptors. Combined FGF2 and 5-HT1A agonist treatment increased the formation of these heterocomplexes and the facilitatory allosteric receptor-receptor interactions within them led to the enhancement of FGFR1 signaling and was associated with the development of antidepressant effects. We discuss these findings with regard to a theory of motifs critically involved in these interactions and suggest that these complexes represent novel targets for antidepressants.
Collapse
Affiliation(s)
| | - Alexander O Tarakanov
- Russian Academy of Sciences, Saint Petersburg Institute for Informatics and Automation, Saint Petersburg, Russia
| | - Kjell Fuxe
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, 17177 Stockholm, Sweden.
| |
Collapse
|
24
|
Goodman T, Hajihosseini MK. Hypothalamic tanycytes-masters and servants of metabolic, neuroendocrine, and neurogenic functions. Front Neurosci 2015; 9:387. [PMID: 26578855 PMCID: PMC4624852 DOI: 10.3389/fnins.2015.00387] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 10/05/2015] [Indexed: 11/13/2022] Open
Abstract
There is a resurgent interest in tanycytes, a radial glial-like cell population occupying the floor and ventro-lateral walls of the third ventricle (3V). Tanycytes reside in close proximity to hypothalamic neuronal nuclei that regulate appetite and energy expenditure, with a subset sending projections into these nuclei. Moreover, tanycytes are exposed to 3V cerebrospinal fluid and have privileged access to plasma metabolites and hormones, through fenestrated capillaries. Indeed, some tanycytes act as conduits for trafficking of these molecules into the brain parenchyma. Tanycytes can also act as neural stem/progenitor cells, supplying the postnatal and adult hypothalamus with new neurons. Collectively, these findings suggest that tanycytes regulate and integrate important trophic and metabolic processes and possibly endow functional malleability to neuronal circuits of the hypothalamus. Hence, manipulation of tanycyte biology could provide a valuable tool for modulating hypothalamic functions such as energy uptake and expenditure in order to tackle prevalent eating disorders such as obesity and anorexia.
Collapse
Affiliation(s)
- Timothy Goodman
- School of Biological Sciences, University of East Anglia Norwich, UK
| | | |
Collapse
|
25
|
Borroto-Escuela DO, Narvaez M, Pérez-Alea M, Tarakanov AO, Jiménez-Beristain A, Mudó G, Agnati LF, Ciruela F, Belluardo N, Fuxe K. Evidence for the existence of FGFR1-5-HT1A heteroreceptor complexes in the midbrain raphe 5-HT system. Biochem Biophys Res Commun 2014; 456:489-93. [PMID: 25485703 DOI: 10.1016/j.bbrc.2014.11.112] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Accepted: 11/28/2014] [Indexed: 11/25/2022]
Abstract
The ascending midbrain 5-HT neurons known to contain 5-HT1A autoreceptors may be dysregulated in depression due to a reduced trophic support. With in situ proximity ligation assay (PLA) and supported by co-location of the FGFR1 and 5-HT1A immunoreactivities in midbrain raphe 5-HT cells, evidence for the existence of FGFR1-5-HT1A heteroreceptor complexes were obtained in the dorsal and median raphe nuclei of the Sprague-Dawley rat. Their existence in the rat medullary raphe RN33B cell cultures was also established. After combined FGF-2 and 8-OH-DPAT treatment, a marked and significant increase in PLA positive clusters was found in the RN33B cells. Similar results were reached upon coactivation by agonists in HEK293T cells using the Fluorescent Resonance Energy Transfer (FRET) technique resulting in increased FRETmax and reduced FRET50 values. The heteroreceptor complex formation was dependent on TMV of the 5-HT1A receptor since it was blocked by incubation with TMV but not with TMII. Taken together, the 5-HT1A autoreceptors by being recruited into a FGFR1-5-HT1A heteroreceptor complex in the midbrain raphe 5-HT nerve cells may develop a novel function, namely a trophic role in many midbrain 5-HT neuron systems originating from the dorsal and medianus raphe nuclei.
Collapse
Affiliation(s)
| | - Manuel Narvaez
- Department of Physiology, School of Medicine, University of Málaga, Spain.
| | - Mileidys Pérez-Alea
- Lab Animal Models and Cancer Laboratory Anatomy Pathology Program, Institut de Recerca Vall d'Hebron, 08035 Barcelona, Spain.
| | - Alexander O Tarakanov
- Russian Academy of Sciences, St. Petersburg Institute for Informatics and Automation, Saint Petersburg, Russia.
| | | | - Giuseppa Mudó
- Department of Experimental Biomedicine and Clinical Neurosciences, Laboratory of Molecular Neurobiology, University of Palermo, Palermo, Italy.
| | - Luigi F Agnati
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | - Francisco Ciruela
- Unitat de Farmacologia, Departament Patologia i Terapèutica Experimental, Universitat de Barcelona, Spain.
| | - Natale Belluardo
- Department of Experimental Biomedicine and Clinical Neurosciences, Laboratory of Molecular Neurobiology, University of Palermo, Palermo, Italy.
| | - Kjell Fuxe
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
26
|
Anatomical, molecular and pathological consideration of the circumventricular organs. Neurochirurgie 2014; 61:90-100. [PMID: 24974365 DOI: 10.1016/j.neuchi.2013.04.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Revised: 04/15/2013] [Accepted: 04/23/2013] [Indexed: 01/25/2023]
Abstract
BACKGROUND AND PURPOSE Circumventricular organs (CVOs) are a diverse group of specialised structures characterized by peculiar vascular and position around the third and fourth ventricles of the brain. In humans, these organs are present during the fetal period and some become vestigial after birth. Some, such as the pineal gland (PG), subcommissural organ (SCO) and organum vasculosum of the lamina terminalis (OVLT), which are located around the third ventricle, might be the site of origin of periventricular tumours. In contrast to humans, CVOs are present in the adult rat and can be dissected by laser capture microdissection (LCM). METHODS In this study, we used LCM and microarrays to analyse the transcriptomes of three CVOs, the SCO, the subfornical organ (SFO) and the PG and the third ventricle ependyma of the adult rat, in order to better characterise these organs at the molecular level. Furthermore, an immunohistochemical study of Claudin-3 (CLDN3), a membrane protein involved in forming cellular tight junctions, was performed at the level of the SCO. RESULTS This study highlighted some potentially new or already described specific markers of these structures as Erbb2 and Col11a1 in ependyma, Epcam and CLDN3 in the SCO, Ren1 and Slc22a3 in the SFO and Tph, Anat and Asmt in the PG. Moreover, we found that CLDN3 expression was restricted to the apical pole of ependymocytes in the SCO.
Collapse
|
27
|
Mäkelä J, Tselykh TV, Maiorana F, Eriksson O, Do HT, Mudò G, Korhonen LT, Belluardo N, Lindholm D. Fibroblast growth factor-21 enhances mitochondrial functions and increases the activity of PGC-1α in human dopaminergic neurons via Sirtuin-1. SPRINGERPLUS 2014; 3:2. [PMID: 25932355 PMCID: PMC4409609 DOI: 10.1186/2193-1801-3-2] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 12/30/2013] [Indexed: 12/18/2022]
Abstract
Mitochondrial dysfunctions accompany several neurodegenerative disorders and contribute to disease pathogenesis among others in Parkinson’s disease (PD). Peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α) is a major regulator of mitochondrial functions and biogenesis, and was suggested as a therapeutic target in PD. PGC-1α is regulated by both transcriptional and posttranslational events involving also the action of growth factors. Fibroblast growth factor-21 (FGF21) is a regulator of glucose and fatty acid metabolism in the body but little is known about its action in the brain. We show here that FGF21 increased the levels and activity of PGC-1α and elevated mitochondrial antioxidants in human dopaminergic cells in culture. The activation of PGC-1α by FGF21 occurred via the NAD+-dependent deacetylase Sirtuin-1 (SIRT1) subsequent to an increase in the enzyme, nicotinamide phosphoribosyltransferase (Nampt). FGF21 also enhanced mitochondrial respiratory capacity in human dopaminergic neurons as shown in real-time analyses of living cells. FGF21 is present in the brain including midbrain and is expressed by glial cells in culture. These results show that FGF21 activates PGC-1α and increases mitochondrial efficacy in human dopaminergic neurons suggesting that FGF21 could potentially play a role in dopaminergic neuron viability and in PD.
Collapse
Affiliation(s)
- Johanna Mäkelä
- Institute of Biomedicine/Biochemistry and Developmental Biology, University of Helsinki, Haartmaninkatu 8, FIN-00290 Helsinki, Finland ; Minerva Foundation Institute for Medical Research, Biomedicum-2, FIN-00290 Helsinki, Finland
| | - Timofey V Tselykh
- Institute of Biomedicine/Biochemistry and Developmental Biology, University of Helsinki, Haartmaninkatu 8, FIN-00290 Helsinki, Finland ; Minerva Foundation Institute for Medical Research, Biomedicum-2, FIN-00290 Helsinki, Finland
| | - Francesca Maiorana
- Institute of Biomedicine/Biochemistry and Developmental Biology, University of Helsinki, Haartmaninkatu 8, FIN-00290 Helsinki, Finland ; Department of Experimental Biomedicine and Clinical Neuroscience, Division of Human Physiology, University of Palermo, Corso Tukory 129, I-90134 Palermo, Italy
| | - Ove Eriksson
- Institute of Biomedicine/Biochemistry and Developmental Biology, University of Helsinki, Haartmaninkatu 8, FIN-00290 Helsinki, Finland
| | - Hai Thi Do
- Institute of Biomedicine/Biochemistry and Developmental Biology, University of Helsinki, Haartmaninkatu 8, FIN-00290 Helsinki, Finland ; Minerva Foundation Institute for Medical Research, Biomedicum-2, FIN-00290 Helsinki, Finland
| | - Giuseppa Mudò
- Department of Experimental Biomedicine and Clinical Neuroscience, Division of Human Physiology, University of Palermo, Corso Tukory 129, I-90134 Palermo, Italy
| | - Laura T Korhonen
- Institute of Biomedicine/Biochemistry and Developmental Biology, University of Helsinki, Haartmaninkatu 8, FIN-00290 Helsinki, Finland ; Minerva Foundation Institute for Medical Research, Biomedicum-2, FIN-00290 Helsinki, Finland
| | - Natale Belluardo
- Department of Experimental Biomedicine and Clinical Neuroscience, Division of Human Physiology, University of Palermo, Corso Tukory 129, I-90134 Palermo, Italy
| | - Dan Lindholm
- Institute of Biomedicine/Biochemistry and Developmental Biology, University of Helsinki, Haartmaninkatu 8, FIN-00290 Helsinki, Finland ; Minerva Foundation Institute for Medical Research, Biomedicum-2, FIN-00290 Helsinki, Finland
| |
Collapse
|
28
|
Song JA, Koo BK, Chong SH, Kwak J, Ryu HB, Nguyen MT, Vu TTT, Jeong B, Kim SW, Choe H. Expression and purification of biologically active human FGF2 containing the b'a' domains of human PDI in Escherichia coli. Appl Biochem Biotechnol 2013; 170:67-80. [PMID: 23471584 DOI: 10.1007/s12010-013-0140-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Accepted: 02/13/2013] [Indexed: 01/31/2023]
Abstract
Among the members of the fibroblast growth factor (FGF) family that affect the growth, differentiation, migration, and survival of many cell types, FGF2 is the most abundant in the central nervous system. Because of its wound healing effects, FGF2 has potential as a therapeutic agent. The protein is also added to the culture media to maintain stem cells. Expression and purification procedures for FGF2 that are highly efficient and low cost have been intensively investigated for the past two decades. Our current study focuses on the purification of FGF2 fused with b'a' domains of human protein disulfide isomerase to elevate overexpression, solubility, and stability with a simplified experimental procedure using only ion exchange chromatography, as well as on the confirmation of the biological activity of FGF2 on fibroblast Balb/c 3T3 cells and hippocampal neural cells.
Collapse
Affiliation(s)
- Jung-A Song
- Department of Physiology and Bio-medical Institute of Technology, University of Ulsan College of Medicine, Seoul 138-736, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
Apert syndrome (Acrocephalosyndactyly type I; AS) is a rare but well-known autosomal dominant disorder characterized by craniosynostosis, midface hypoplasia, bony/cutaneous syndactyly of fingers and toes as well as a variety of associated congenital anomalies involving the brain, heart, limbs and other organ systems. We report the case of a fetus with molecularly confirmed Apert syndrome and additional fusion of the thalamic nuclei. Various central nervous system anomalies, have been reported in patients with AS. However, as far as we know cases of fused thalami in Apert syndrome have never been reported so far.
Collapse
Affiliation(s)
- Kathrin Ludwig
- Pathology Unit, Department of Medical Diagnostic Sciences & Special Therapies, Padova, Italy
| | | | | | | | | | | |
Collapse
|
30
|
Huettl RE, Haehl T, Huber AB. Fasciculation and guidance of spinal motor axons in the absence of FGFR2 signaling. PLoS One 2012; 7:e41095. [PMID: 22815929 PMCID: PMC3398880 DOI: 10.1371/journal.pone.0041095] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Accepted: 06/18/2012] [Indexed: 11/18/2022] Open
Abstract
During development, fibroblast growth factors (FGF) are essential for early patterning events along the anterior-posterior axis, conferring positional identity to spinal motor neurons by activation of different Hox codes. In the periphery, signaling through one of four fibroblast growth factor receptors supports the development of the skeleton, as well as induction and maintenance of extremities. In previous studies, FGF receptor 2 (FGFR2) was found to interact with axon bound molecules involved in axon fasciculation and extension, thus rendering this receptor an interesting candidate for the promotion of proper peripheral innervation. However, while the involvement of FGFR2 in limb bud induction has been extensively studied, its role during axon elongation and formation of distinct nervous projections has not been addressed so far. We show here that motor neurons in the spinal cord express FGFR2 and other family members during the establishment of motor connections to the forelimb and axial musculature. Employing a conditional genetic approach to selectively ablate FGFR2 from motor neurons we found that the patterning of motor columns and the expression patterns of other FGF receptors and Sema3A in the motor columns of mutant embryos are not altered. In the absence of FGFR2 signaling, pathfinding of motor axons is intact, and also fasciculation, distal advancement of motor nerves and gross morphology and positioning of axonal projections are not altered. Our findings therefore show that FGFR2 is not required cell-autonomously in motor neurons during the formation of initial motor projections towards limb and axial musculature.
Collapse
Affiliation(s)
- Rosa-Eva Huettl
- Institute of Developmental Genetics, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
| | - Teresa Haehl
- Institute of Developmental Genetics, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
| | - Andrea B. Huber
- Institute of Developmental Genetics, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
- * E-mail:
| |
Collapse
|
31
|
Szathmari A, Champier J, Ghersi-Egea JF, Jouvet A, Watrin C, Wierinckx A, Fèvre Montange M. Molecular characterization of circumventricular organs and third ventricle ependyma in the rat: potential markers for periventricular tumors. Neuropathology 2012; 33:17-29. [PMID: 22537279 DOI: 10.1111/j.1440-1789.2012.01321.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Circumventricular organs (CVOs) are specialized ventricular structures around the third and fourth ventricles of the brain. In humans, these structures are present during the fetal period and some become vestigial after birth. Some of these organs, such as the pineal gland (PG), subcommissural organ (SCO), and organum vasculosum of the lamina terminalis, might be the sites of origin of periventricular tumors, notably pineal parenchymal tumors, papillary tumor of the pineal region and chordoid glioma. In contrast to the situation in humans, CVOs are present in the adult rat and can be dissected by laser capture microdissection (LCM). In this study, we used LCM and microarrays to analyze the transcriptomes of three CVOs, the SCO, the subfornical organ (SFO), and the PG and the third ventricle ependyma in the adult rat, in order to better characterize these organs at the molecular level. Several genes were expressed only, or mainly, in one of these structures, for example, Erbb2 and Col11a1 in the ependyma, Epcam and Claudin-3 (CLDN3) in the SCO, Ren1 and Slc22a3 in the SFO and Tph, Aanat and Asmt in the PG. The expression of these genes in periventricular tumors should be examined as evidence for a possible origin from the CVOs. Furthermore, we performed an immunohistochemical study of CLDN3, a membrane protein involved in forming cellular tight junctions and found that CLDN3 expression was restricted to the apical pole of ependymocytes in the SCO. This microarray study provides new evidence regarding the possible origin of some rare periventricular tumors.
Collapse
Affiliation(s)
- Alexandru Szathmari
- Fac Med RTH Laennec, Inserm U1028, CNRS UMR5292, Centre de Recherche en Neurosciences, Equipe Neurooncologie et Neuroinflammation, Université de Lyon, Lyon, France
| | | | | | | | | | | | | |
Collapse
|
32
|
Knafo S, Venero C, Sánchez-Puelles C, Pereda-Peréz I, Franco A, Sandi C, Suárez LM, Solís JM, Alonso-Nanclares L, Martín ED, Merino-Serrais P, Borcel E, Li S, Chen Y, Gonzalez-Soriano J, Berezin V, Bock E, DeFelipe J, Esteban JA. Facilitation of AMPA receptor synaptic delivery as a molecular mechanism for cognitive enhancement. PLoS Biol 2012; 10:e1001262. [PMID: 22363206 PMCID: PMC3283560 DOI: 10.1371/journal.pbio.1001262] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2011] [Accepted: 01/05/2012] [Indexed: 11/19/2022] Open
Abstract
Cell adhesion molecules and downstream growth factor-dependent signaling are critical for brain development and synaptic plasticity, and they have been linked to cognitive function in adult animals. We have previously developed a mimetic peptide (FGL) from the neural cell adhesion molecule (NCAM) that enhances spatial learning and memory in rats. We have now investigated the cellular and molecular basis of this cognitive enhancement, using biochemical, morphological, electrophysiological, and behavioral analyses. We have found that FGL triggers a long-lasting enhancement of synaptic transmission in hippocampal CA1 neurons. This effect is mediated by a facilitated synaptic delivery of AMPA receptors, which is accompanied by enhanced NMDA receptor-dependent long-term potentiation (LTP). Both LTP and cognitive enhancement are mediated by an initial PKC activation, which is followed by persistent CaMKII activation. These results provide a mechanistic link between facilitation of AMPA receptor synaptic delivery and improved hippocampal-dependent learning, induced by a pharmacological cognitive enhancer.
Collapse
Affiliation(s)
- Shira Knafo
- Centro de Biología Molecular “Severo Ochoa,” Consejo Superior de Investigaciones Científicas (CSIC)/Universidad Autónoma de Madrid, Madrid, Spain
- Instituto Cajal (CSIC), Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - César Venero
- Department of Psychobiology, Universidad Nacional de Educación a Distancia, Madrid, Spain
| | - Cristina Sánchez-Puelles
- Centro de Biología Molecular “Severo Ochoa,” Consejo Superior de Investigaciones Científicas (CSIC)/Universidad Autónoma de Madrid, Madrid, Spain
| | | | - Ana Franco
- Centro Nacional Biotecnología (CSIC), Universidad Autónoma de Madrid, Madrid, Spain
| | - Carmen Sandi
- Brain Mind Institute, Ecole Polytechnique Federale de Lausanne (EPFL), Switzerland
| | - Luz M. Suárez
- Instituto Cajal (CSIC), Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Servicio de Neurobiología-Investigación, Hospital Ramón y Cajal, IRYCIS, Madrid, Spain
| | - José M. Solís
- Servicio de Neurobiología-Investigación, Hospital Ramón y Cajal, IRYCIS, Madrid, Spain
| | - Lidia Alonso-Nanclares
- Instituto Cajal (CSIC), Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Eduardo D. Martín
- Departamento de Ciencias Médicas, Universidad de Castilla-la Mancha, Albacete, Spain
| | - Paula Merino-Serrais
- Instituto Cajal (CSIC), Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Erika Borcel
- Department of Psychobiology, Universidad Nacional de Educación a Distancia, Madrid, Spain
| | - Shizhong Li
- Protein Laboratory, Department of Neuroscience and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Yongshuo Chen
- Protein Laboratory, Department of Neuroscience and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Juncal Gonzalez-Soriano
- Department of Anatomy, Faculty of Veterinary Medicine, Complutense University, Madrid, Spain
| | - Vladimir Berezin
- Protein Laboratory, Department of Neuroscience and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Elisabeth Bock
- Protein Laboratory, Department of Neuroscience and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Javier DeFelipe
- Instituto Cajal (CSIC), Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - José A. Esteban
- Centro de Biología Molecular “Severo Ochoa,” Consejo Superior de Investigaciones Científicas (CSIC)/Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
33
|
Lahti L, Peltopuro P, Piepponen TP, Partanen J. Cell-autonomous FGF signaling regulates anteroposterior patterning and neuronal differentiation in the mesodiencephalic dopaminergic progenitor domain. Development 2012; 139:894-905. [PMID: 22278924 DOI: 10.1242/dev.071936] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The structure and projection patterns of adult mesodiencephalic dopaminergic (DA) neurons are one of the best characterized systems in the vertebrate brain. However, the early organization and development of these nuclei remain poorly understood. The induction of midbrain DA neurons requires sonic hedgehog (Shh) from the floor plate and fibroblast growth factor 8 (FGF8) from the isthmic organizer, but the way in which FGF8 regulates DA neuron development is unclear. We show that, during early embryogenesis, mesodiencephalic neurons consist of two distinct populations: a diencephalic domain, which is probably independent of isthmic FGFs; and a midbrain domain, which is dependent on FGFs. Within these domains, DA progenitors and precursors use partly different genetic programs. Furthermore, the diencephalic DA domain forms a distinct cell population, which also contains non-DA Pou4f1(+) cells. FGF signaling operates in proliferative midbrain DA progenitors, but is absent in postmitotic DA precursors. The loss of FGFR1/2-mediated signaling results in a maturation failure of the midbrain DA neurons and altered patterning of the midbrain floor. In FGFR mutants, the DA domain adopts characteristics that are typical for embryonic diencephalon, including the presence of Pou4f1(+) cells among TH(+) cells, and downregulation of genes typical of midbrain DA precursors. Finally, analyses of chimeric embryos indicate that FGF signaling regulates the development of the ventral midbrain cell autonomously.
Collapse
Affiliation(s)
- Laura Lahti
- Department of Biosciences, Division of Genetics, University of Helsinki, Helsinki, Finland
| | | | | | | |
Collapse
|
34
|
Borroto-Escuela DO, Romero-Fernandez W, Mudó G, Pérez-Alea M, Ciruela F, Tarakanov AO, Narvaez M, Di Liberto V, Agnati LF, Belluardo N, Fuxe K. Fibroblast growth factor receptor 1- 5-hydroxytryptamine 1A heteroreceptor complexes and their enhancement of hippocampal plasticity. Biol Psychiatry 2012; 71:84-91. [PMID: 22035699 DOI: 10.1016/j.biopsych.2011.09.012] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Revised: 09/06/2011] [Accepted: 09/08/2011] [Indexed: 10/15/2022]
Abstract
BACKGROUND The hippocampus and its 5-hydroxytryptamine transmission plays an important role in depression related to its involvement in limbic circuit plasticity. METHODS The analysis was made with bioluminescence resonance energy transfer, co-immunoprecipitation, in situ proximity ligation assay, binding assay, in cell western and the forced swim test. RESULTS Using bioluminescence resonance energy transfer analysis, fibroblast growth factor receptor 1 (FGFR1)-5-hydroxytryptamine 1A (5-HT1A) receptor complexes have been demonstrated and their specificity and agonist modulation characterized. Their presence based on co-immunoprecipitation and proximity ligation assay has also been indicated in hippocampal cultures and rat dorsal hippocampal formation showing a neuronal location. In vitro assays on extracellular signal-regulated kinases 1 and 2 phosphorylation have shown synergistic increases in signaling on coactivation with fibroblast growth factor 2 (FGF2) and a 5-HT1A agonist, and dependent on the heteroreceptor interface. In vitro and in vivo studies also revealed a 5-HT1A agonist induced phosphorylation of FGFR1 and extracellular signal-regulated kinase 1/2 in rat hippocampus without changing FGF2 levels. Co-activation of the heteroreceptor also resulted in synergistic increases in extensions of PC12 cells and neurite densities and protrusions in primary hippocampal cultures dependent on the receptor interface. The combined acute and repeated intracerebroventricular treatment with FGF2 and 8-OH-DPAT was found to produce evidence of highly significant antidepressant actions in the forced swim test. CONCLUSIONS The findings indicate that neurotrophic and antidepressant effects of 5-HT in brain may, in part, be mediated by activation of the 5-HT1A receptor protomer in the hippocampal FGFR1-5-HT1A receptor complex enhancing the FGFR1 signaling.
Collapse
|
35
|
Lichtenstein MP, Madrigal JLM, Pujol A, Galea E. JNK/ERK/FAK mediate promigratory actions of basic fibroblast growth factor in astrocytes via CCL2 and COX2. Neurosignals 2011; 20:86-102. [PMID: 22189091 DOI: 10.1159/000330805] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Accepted: 07/07/2011] [Indexed: 12/31/2022] Open
Abstract
While the role of cytokines in causing pro- and anti-inflammatory cascades in the brain and that of chemokines in promoting chemotaxis is well recognized, the immunomodulatory actions of neurotrophins released during brain injury remains largely undetermined. This knowledge gap affects basic fibroblast growth factor (FGF2), which in the brain is mainly produced by astrocytes and characteristically upregulated in reactive astrocytes. The goal of this study was to characterize the inflammatory actions of FGF2 in astrocytes using primary cultures. We report that FGF2 induced the upregulation of monocyte chemoattractant protein (CCL2) and cyclo-oxygenase type 2 (COX2), and the inhibition of lipopolysaccharide-elicited ICAM1 upregulation. The effects of FGF2 were: (i) dependent on gene transcription as revealed by the concomitant regulation of CCL2 or ICAM1 mRNAs; (ii) mediated by the FGF2 receptor type 2; (iii) dependent on ERK, JNK and FAK, and (iv) NF-κB-independent. FGF2 also caused accelerated wound closure dependent on CCL2, COX2, ERK, JNK and FAK in a scratch assay. We conclude that the signaling network triggered by FGF2 in astrocytes converged into accelerating directed motion. It follows that astrocyte migration to injury sites may be a key factor in the repair mechanisms orchestrated by FGF2.
Collapse
|
36
|
Kraev I, Henneberger C, Rossetti C, Conboy L, Kohler LB, Fantin M, Jennings A, Venero C, Popov V, Rusakov D, Stewart MG, Bock E, Berezin V, Sandi C. A peptide mimetic targeting trans-homophilic NCAM binding sites promotes spatial learning and neural plasticity in the hippocampus. PLoS One 2011; 6:e23433. [PMID: 21887252 PMCID: PMC3160849 DOI: 10.1371/journal.pone.0023433] [Citation(s) in RCA: 21] [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: 04/06/2011] [Accepted: 07/17/2011] [Indexed: 01/10/2023] Open
Abstract
The key roles played by the neural cell adhesion molecule (NCAM) in plasticity and cognition underscore this membrane protein as a relevant target to develop cognitive-enhancing drugs. However, NCAM is a structurally and functionally complex molecule with multiple domains engaged in a variety of actions, which raise the question as to which NCAM fragment should be targeted. Synthetic NCAM mimetic peptides that mimic NCAM sequences relevant to specific interactions allow identification of the most promising targets within NCAM. Recently, a decapeptide ligand of NCAM—plannexin, which mimics a homophilic trans-binding site in Ig2 and binds to Ig3—was developed as a tool for studying NCAM's trans-interactions. In this study, we investigated plannexin's ability to affect neural plasticity and memory formation. We found that plannexin facilitates neurite outgrowth in primary hippocampal neuronal cultures and improves spatial learning in rats, both under basal conditions and under conditions involving a deficit in a key plasticity-promoting posttranslational modification of NCAM, its polysialylation. We also found that plannexin enhances excitatory synaptic transmission in hippocampal area CA1, where it also increases the number of mushroom spines and the synaptic expression of the AMPAR subunits GluA1 and GluA2. Altogether, these findings provide compelling evidence that plannexin is an important facilitator of synaptic functional, structural and molecular plasticity in the hippocampal CA1 region, highlighting the fragment in NCAM's Ig3 module where plannexin binds as a novel target for the development of cognition-enhancing drugs.
Collapse
Affiliation(s)
- Igor Kraev
- Department of Life Sciences, The Open University, Milton Keynes, United Kingdom
| | - Christian Henneberger
- Department of Clinical and Experimental Epilepsy, Institute of Neurology, University College London (UCL), London, United Kingdom
| | - Clara Rossetti
- Laboratory of Behavioral Genetics, Brain Mind Institute, EPFL, Lausanne, Switzerland
| | - Lisa Conboy
- Laboratory of Behavioral Genetics, Brain Mind Institute, EPFL, Lausanne, Switzerland
| | - Lene B. Kohler
- Protein Laboratory, Department of Neuroscience and Pharmacology, Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Martina Fantin
- Laboratory of Behavioral Genetics, Brain Mind Institute, EPFL, Lausanne, Switzerland
| | - Alistair Jennings
- Department of Clinical and Experimental Epilepsy, Institute of Neurology, University College London (UCL), London, United Kingdom
| | - Cesar Venero
- Department of Psychobiology, UNED, Ciudad Universitaria, Madrid, Spain
| | - Victor Popov
- Department of Life Sciences, The Open University, Milton Keynes, United Kingdom
| | - Dmitri Rusakov
- Department of Clinical and Experimental Epilepsy, Institute of Neurology, University College London (UCL), London, United Kingdom
| | - Michael G. Stewart
- Department of Life Sciences, The Open University, Milton Keynes, United Kingdom
- * E-mail: (CS); (MGS)
| | - Elisabeth Bock
- Protein Laboratory, Department of Neuroscience and Pharmacology, Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Vladimir Berezin
- Protein Laboratory, Department of Neuroscience and Pharmacology, Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Carmen Sandi
- Laboratory of Behavioral Genetics, Brain Mind Institute, EPFL, Lausanne, Switzerland
- * E-mail: (CS); (MGS)
| |
Collapse
|
37
|
Zechel S, Werner S, Unsicker K, von Bohlen und Halbach O. Expression and Functions of Fibroblast Growth Factor 2 (FGF-2) in Hippocampal Formation. Neuroscientist 2010; 16:357-73. [DOI: 10.1177/1073858410371513] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Among the 23 members of the fibroblast growth factor (FGF) family, FGF-2 is the most abundant one in the central nervous system. Its impact on neural cells has been profoundly investigated by in vitro and in vivo studies as well as by gene knockout analyses during the past 2 decades. Key functions of FGF-2 in the nervous system include roles in neurogenesis, promotion of axonal growth, differentiation in development, and maintenance and plasticity in adulthood. From a clinical perspective, its prominent role for the maintenance of lesioned neurons (e.g., ischemia and following transection of fiber tracts) is of particular relevance. In the unlesioned brain, FGF-2 is involved in synaptic plasticity and processes attributed to learning and memory. The focus of this review is on the expression of FGF-2 and its receptors in the hippocampal formation and the physiological and pathophysiological roles of FGF-2 in this region during development and adulthood.
Collapse
Affiliation(s)
- Sabrina Zechel
- Division of Molecular Neurobiology, Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Sandra Werner
- Department of Molecular Embryology, Institute of Anatomy & Cell Biology, University of Freiburg, Freiburg, Germany
| | - Klaus Unsicker
- Department of Molecular Embryology, Institute of Anatomy & Cell Biology, University of Freiburg, Freiburg, Germany
| | | |
Collapse
|
38
|
Flores J, Galan-Rodriguez B, Rojo A, Ramiro-Fuentes S, Cuadrado A, Fernandez-Espejo E. Fibroblast growth factor-1 within the ventral tegmental area participates in motor sensitizing effects of morphine. Neuroscience 2010; 165:198-211. [DOI: 10.1016/j.neuroscience.2009.10.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2009] [Revised: 09/03/2009] [Accepted: 10/03/2009] [Indexed: 11/16/2022]
|
39
|
The FGF-2/FGFRs neurotrophic system promotes neurogenesis in the adult brain. J Neural Transm (Vienna) 2009; 116:995-1005. [PMID: 19291360 DOI: 10.1007/s00702-009-0207-z] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2008] [Accepted: 02/19/2009] [Indexed: 01/03/2023]
Abstract
Neurogenesis occurs in two regions of the adult brain, namely, the subventricular zone (SVZ) throughout the wall of the lateral ventricle and the subgranular zone (SGZ) of the dentate gyrus (DG) in hippocampal formation. Adult neurogenesis requires several neurotrophic factors to sustain and regulate the proliferation and differentiation of the adult stem cell population. In the present review, we examine the cellular and functional aspects of a trophic system mediated by fibroblast growth factor-2 (FGF-2) and its receptors (FGFRs) related to neurogenesis in the SVZ and SGZ of the adult rat brain. In the SVZ, FGF-2 is expressed in GFAP-positive cells of SVZ but is not present in proliferating precursor cells, which instead express FGFR-1 and FGFR-2, but not FGFR-3 mRNA, although expressed in the SVZ, and FGFR-4. Therefore, it seems that in the SVZ FGF-2 may be released by GFAP-positive cells, different from the precursor cell lineage, and via volume transmission it reaches the proliferating precursor cells. FGFR-1 mRNA is also expressed in the SGZ and is localized in BrdU-labeled precursor cells, whereas FGFR-2 and FGFR-3 mRNA, although expressed in the SGZ, are not located within proliferating precursor cells. An aged-related decline of proliferating precursor cells in the SVZ and DG of old rats has been well documented, and there is the suggestion that in part it could be the consequence of alterations in growth factor expression levels. Thus, the old precursors may respond to growth factors, suggesting that during aging the basic components for neuronal precursor cell proliferation are retained and the capacity to increase neurogenesis after appropriate stimulation is still preserved. In conclusion, the trophic system mediated by FGF-2 and its receptors contributes to create an important micro-environmental niche that promotes neurogenesis in the adult and aged brain.
Collapse
|
40
|
Zechel S, Unsicker K, von Bohlen und Halbach O. Fibroblast growth factor-2 deficiency affects hippocampal spine morphology, but not hippocampal catecholaminergic or cholinergic innervation. Dev Dyn 2009; 238:343-50. [DOI: 10.1002/dvdy.21839] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
41
|
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]
|
42
|
Frinchi M, Bonomo A, Trovato-Salinaro A, Condorelli DF, Fuxe K, Spampinato MG, Mudò G. Fibroblast growth factor-2 and its receptor expression in proliferating precursor cells of the subventricular zone in the adult rat brain. Neurosci Lett 2008; 447:20-5. [PMID: 18835325 DOI: 10.1016/j.neulet.2008.09.059] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2008] [Revised: 09/11/2008] [Accepted: 09/13/2008] [Indexed: 12/13/2022]
Abstract
Several findings have suggested the existence in the subventricular zone (SVZ) from sagittal sections of adult rat brain of a trophic mechanism, mediated by fibroblast growth factor-2 (FGF-2) and its multiple high-affinity FGF receptors (FGFRs), regulating neurogenesis mainly by controlling precursor cell proliferation. However, no clear data are available on the expression of FGF-2 and FGFRs in proliferating precursor cells of the SVZ. To address these questions we examined FGF-2 mRNA and its FGFR mRNA expression in proliferating precursor cells of the SVZ by using a double labeling procedure, combining in situ hybridization for FGF-2 and its FGFR mRNA with immunohistochemistry for bromodeoxyuridine (BrdU), a marker for proliferating cells. The results showed that FGFR1 and FGFR2 mRNAs were expressed in BrdU-labeled proliferating precursor cells, whereas FGFR3 and FGF-2 mRNAs were not, suggesting that in the SVZ the proliferating precursor cells express FGFR1 or FGFR2 and they may respond to FGF-2 released by non-proliferating cells. The FGFR4 mRNA was not found expressed in the SVZ. In the future, by identifying the cell types expressing FGFRs, it will be possible to gain insight into the functional activity of FGF2 within the SVZ.
Collapse
MESH Headings
- Adult Stem Cells/metabolism
- Animals
- Brain/anatomy & histology
- Bromodeoxyuridine/metabolism
- Cell Proliferation
- Cerebral Ventricles/cytology
- Fibroblast Growth Factor 2/genetics
- Fibroblast Growth Factor 2/metabolism
- Gene Expression/physiology
- Male
- RNA, Messenger/metabolism
- Rats
- Rats, Wistar
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Receptor, Fibroblast Growth Factor, Type 1/metabolism
- Receptor, Fibroblast Growth Factor, Type 2/genetics
- Receptor, Fibroblast Growth Factor, Type 2/metabolism
- Receptor, Fibroblast Growth Factor, Type 3/genetics
- Receptor, Fibroblast Growth Factor, Type 3/metabolism
Collapse
Affiliation(s)
- Monica Frinchi
- Department of Experimental Medicine, Division of Human Physiology, University of Palermo, Palermo, Italy
| | | | | | | | | | | | | |
Collapse
|
43
|
Chojnacki A, Kelly JJP, Hader W, Weiss S. Distinctions between fetal and adult human platelet-derived growth factor-responsive neural precursors. Ann Neurol 2008; 64:127-42. [DOI: 10.1002/ana.21421] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
44
|
Adrenalectomy counteracts the local modulation of astroglial fibroblast growth factor system without interfering with the pattern of 6-OHDA-induced dopamine degeneration in regions of the ventral midbrain. Brain Res 2008; 1190:23-38. [PMID: 18086466 DOI: 10.1016/j.brainres.2007.11.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2007] [Revised: 10/29/2007] [Accepted: 11/07/2007] [Indexed: 11/21/2022]
|
45
|
Belluardo N, Mudo' G, Bonomo A, Di Liberto V, Frinchi M, Fuxe K. Nicotine-induced fibroblast growth factor-2 restores the age-related decline of precursor cell proliferation in the subventricular zone of rat brain. Brain Res 2007; 1193:12-24. [PMID: 18190895 DOI: 10.1016/j.brainres.2007.11.069] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2007] [Revised: 11/17/2007] [Accepted: 11/26/2007] [Indexed: 01/27/2023]
Abstract
Precursor cell proliferation is present in the subventricular zone (SVZ) of the lateral ventricles and the subgranular zone (SGZ) of the dentate gyrus of the hippocampus of adult rat and persists during aging although at reduced levels. Previous studies have shown that acute intermittent nicotine treatment significantly increases fibroblast growth factor-2 (FGF-2) expression in several brain regions of aged rats. The aim of the present investigation was to test the hypothesis that nicotine-induced expression of FGF-2 may restore the age-related decline of precursor cell proliferation. It was first demonstrated that nicotine treatment increases both mRNA and protein FGF-2 in the SVZ of aged male rats (18 months old). The effect of nicotine on precursor cell proliferation in the SVZ was studied by i.p. injection of 5-bromo-2'-deoxyuridine (BrdU) 40 mg/kg to label dividing cells. The nicotine treatment was found to significantly enhance precursor cell proliferation in the SVZ. This increase was sufficiently large to restore the age-related decline of proliferating precursor cells observed in aged rats to that found in young adult rats (3 months old). FGF-2 was expressed in GFAP-positive cells and may act via its receptor FGFR1 that was found expressed in nestin-positive cells of the SVZ. The data obtained demonstrated that the age-related decline of precursor cell proliferation may be counteracted by activating a trophic mechanism mediated by FGF-2.
Collapse
Affiliation(s)
- Natale Belluardo
- Department of Experimental Medicine, Division of Human Physiology, Laboratory of Molecular Neurobiology, University of Palermo, corso Tukory 129, 90134 Palermo, Italy.
| | | | | | | | | | | |
Collapse
|
46
|
Rai KS, Hattiangady B, Shetty AK. Enhanced production and dendritic growth of new dentate granule cells in the middle-aged hippocampus following intracerebroventricular FGF-2 infusions. Eur J Neurosci 2007; 26:1765-79. [PMID: 17883411 DOI: 10.1111/j.1460-9568.2007.05820.x] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Declined production and diminished dendritic growth of new dentate granule cells in the middle-aged and aged hippocampus are correlated with diminished concentration of fibroblast growth factor-2 (FGF-2). This study examined whether increased FGF-2 concentration in the milieu boosts both production and dendritic growth of new dentate granule cells in the middle-aged hippocampus. The FGF-2 or vehicle was infused into the posterior lateral ventricle of middle-aged Fischer (F)344 rats for 2 weeks using osmotic minipumps. New cells born during the first 12 days of infusions were labeled via daily intraperitoneal injections of 5'-bromodeoxyuridine (BrdU) and analysed at 10 days after the last BrdU injection. Measurement of BrdU(+) cells revealed a considerably enhanced number of new cells in the subgranular zone (SGZ) and granule cell layer (GCL) of the dentate gyrus (DG) ipsilateral to FGF-2 infusions. Characterization of beta-III tubulin(+) neurons among newly born cells suggested an increased addition of new neurons to the SGZ/GCL ipsilateral to FGF-2 infusions. Quantification of DG neurogenesis at 8 days post-infusions via doublecortin (DCX) immunostaining also revealed the presence of an enhanced DG neurogenesis ipsilateral to FGF-2 infusions. Furthermore, DCX(+) neurons in FGF-2-infused rats exhibited enhanced dendritic growth compared with their counterparts in vehicle-infused rats. Thus, subchronic infusion of FGF-2 is efficacious for stimulating an enhanced DG neurogenesis from neural stem/progenitor cells in the middle-aged hippocampus. As dentate neurogenesis is important for hippocampal-dependent learning and memory and DG long-term potentiation, strategies that maintain increased FGF-2 concentration during ageing may be beneficial for thwarting some of the age-related cognitive impairments.
Collapse
Affiliation(s)
- Kiranmai S Rai
- Department of Surgery (Neurosurgery), Duke University Medical Center, Durham, NC 27710, USA
| | | | | |
Collapse
|
47
|
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
|
48
|
Fuxe K, Dahlström A, Höistad M, Marcellino D, Jansson A, Rivera A, Diaz-Cabiale Z, Jacobsen K, Tinner-Staines B, Hagman B, Leo G, Staines W, Guidolin D, Kehr J, Genedani S, Belluardo N, Agnati LF. From the Golgi–Cajal mapping to the transmitter-based characterization of the neuronal networks leading to two modes of brain communication: Wiring and volume transmission. ACTA ACUST UNITED AC 2007; 55:17-54. [PMID: 17433836 DOI: 10.1016/j.brainresrev.2007.02.009] [Citation(s) in RCA: 182] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2007] [Revised: 02/21/2007] [Accepted: 02/27/2007] [Indexed: 10/23/2022]
Abstract
After Golgi-Cajal mapped neural circuits, the discovery and mapping of the central monoamine neurons opened up for a new understanding of interneuronal communication by indicating that another form of communication exists. For instance, it was found that dopamine may be released as a prolactin inhibitory factor from the median eminence, indicating an alternative mode of dopamine communication in the brain. Subsequently, the analysis of the locus coeruleus noradrenaline neurons demonstrated a novel type of lower brainstem neuron that monosynaptically and globally innervated the entire CNS. Furthermore, the ascending raphe serotonin neuron systems were found to globally innervate the forebrain with few synapses, and where deficits in serotonergic function appeared to play a major role in depression. We propose that serotonin reuptake inhibitors may produce antidepressant effects through increasing serotonergic neurotrophism in serotonin nerve cells and their targets by transactivation of receptor tyrosine kinases (RTK), involving direct or indirect receptor/RTK interactions. Early chemical neuroanatomical work on the monoamine neurons, involving primitive nervous systems and analysis of peptide neurons, indicated the existence of alternative modes of communication apart from synaptic transmission. In 1986, Agnati and Fuxe introduced the theory of two main types of intercellular communication in the brain: wiring and volume transmission (WT and VT). Synchronization of phasic activity in the monoamine cell clusters through electrotonic coupling and synaptic transmission (WT) enables optimal VT of monoamines in the target regions. Experimental work suggests an integration of WT and VT signals via receptor-receptor interactions, and a new theory of receptor-connexin interactions in electrical and mixed synapses is introduced. Consequently, a new model of brain function must be built, in which communication includes both WT and VT and receptor-receptor interactions in the integration of signals. This will lead to the unified execution of information handling and trophism for optimal brain function and survival.
Collapse
Affiliation(s)
- Kjell Fuxe
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
Achondroplasia is the most common form of short limb dwarfism in human beings, affecting more than 250,000 individuals worldwide. More than 95% of patients have the same point mutation in the gene for fibroblast growth factor receptor 3 (FGFR3) and more than 80% of these are new mutations. The mutation, which causes gain of FGFR3 function, affects many tissues, most strikingly the cartilaginous growth plate in the growing skeleton, leading to a variety of manifestations and complications. The biology of FGFR3 and the molecular and cellular consequences of the achondroplasia mutation are being elucidated, providing a more complete understanding of the disorder and a basis for future treatments targeted directly at relevant pathogenetic pathways. Furthermore, the natural history of the condition, which has been well delineated in childhood and adolescence, is being defined more fully in adults with achondroplasia; most of the serious complications can be modified favourably or prevented by anticipation and early treatment. Possible future treatments include chemical inhibition of receptor signalling, antibody blockade of receptor activation, and alteration of pathways that modulate the downstream propagation of FGFR3 signals.
Collapse
Affiliation(s)
- William A Horton
- Research Center, Shriners Hospital for Children and Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA.
| | - Judith G Hall
- Departments of Pediatrics and Medical Genetics, University of British Columbia and Department of Pediatrics, British Columbia Children's Hospital, Vancouver, BC, Canada
| | - Jacqueline T Hecht
- Department of Pediatrics, University of Texas Medical School at Houston, Houston, TX, USA
| |
Collapse
|
50
|
Grothe C, Timmer M. The physiological and pharmacological role of basic fibroblast growth factor in the dopaminergic nigrostriatal system. ACTA ACUST UNITED AC 2007; 54:80-91. [PMID: 17229467 DOI: 10.1016/j.brainresrev.2006.12.001] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2006] [Revised: 11/28/2006] [Accepted: 12/11/2006] [Indexed: 12/24/2022]
Abstract
Basic fibroblast growth factor (FGF-2) is a physiological relevant neurotrophic factor in the nigrostriatal system and hence a promising candidate for the establishment of alternative therapeutic strategies in Parkinson's disease. FGF-2 and its high-affinity receptors (FGFR) display an expression in the developing, postnatal, and adult substantia nigra (SN) and in the striatum. Exogenous application promoted survival, neurite outgrowth and protection from neurotoxin-induced death of dopaminergic (DA) neurons both in vitro and in vivo. In animal models of Parkinson's disease, co-transplantation of fetal DA cells with FGF-2 expressing cells increased survival and functional integration of the grafted DA neurons resulting in improved behavioral performance. Analyzing the physiological function of the endogenous FGF-2 system during development and after neurotoxin-induced lesion revealed for the DA neurons of the SNpc a dependence on FGFR3 signaling during development. In addition, in the absence of FGF-2 an increased number of DA neurons was found, whereas enhanced levels of FGF-2 resulted in a reduced DA cell density. Following neurotoxin-induced lesion of DA neurons, FGF-2-deleted mice displayed a higher extent of DA neuron death whereas in FGF-2 overexpressing mice more DA neurons were protected. According to the data, FGF-2 seems to promote DA neuron survival via FGFR3 during development, whereas absence of this ligand could be compensated by other members of the FGF family. In contrast, in the adult organism, FGF-2 cannot be compensated by other factors under lesion conditions suggesting a central role for this molecule in the nigrostriatal system.
Collapse
Affiliation(s)
- Claudia Grothe
- Department of Neuroanatomy, Hannover Medical School, OE 4140, Center for Systems Neuroscience Hannover (ZSN), Carl-Neuberg-Strasse 1, 30625 Hannover, Germany.
| | | |
Collapse
|