1
|
Dhiman S, Manoj N, Liput M, Sangwan A, Diehl J, Balcerak A, Sudhakar S, Augustyniak J, Jornet JM, Bae Y, Stachowiak EK, Dutta A, Stachowiak MK. Systems Genome: Coordinated Gene Activity Networks, Recurring Coordination Modules, and Genome Homeostasis in Developing Neurons. Int J Mol Sci 2024; 25:5647. [PMID: 38891836 PMCID: PMC11171963 DOI: 10.3390/ijms25115647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 05/02/2024] [Accepted: 05/10/2024] [Indexed: 06/21/2024] Open
Abstract
As human progenitor cells differentiate into neurons, the activities of many genes change; these changes are maintained within a narrow range, referred to as genome homeostasis. This process, which alters the synchronization of the entire expressed genome, is distorted in neurodevelopmental diseases such as schizophrenia. The coordinated gene activity networks formed by altering sets of genes comprise recurring coordination modules, governed by the entropy-controlling action of nuclear FGFR1, known to be associated with DNA topology. These modules can be modeled as energy-transferring circuits, revealing that genome homeostasis is maintained by reducing oscillations (noise) in gene activity while allowing gene activity changes to be transmitted across networks; this occurs more readily in neuronal committed cells than in neural progenitors. These findings advance a model of an "entangled" global genome acting as a flexible, coordinated homeostatic system that responds to developmental signals, is governed by nuclear FGFR1, and is reprogrammed in disease.
Collapse
Affiliation(s)
- Siddhartha Dhiman
- Department of Biomedical Engineering, University at Buffalo, Buffalo, NY 14228, USA; (S.D.); (A.D.)
| | - Namya Manoj
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14228, USA; (N.M.); (M.L.); (J.D.); (A.B.); (S.S.); (J.A.); (Y.B.); (E.K.S.)
| | - Michal Liput
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14228, USA; (N.M.); (M.L.); (J.D.); (A.B.); (S.S.); (J.A.); (Y.B.); (E.K.S.)
- Mossakowski Medical Research Center, Stem Cell Bioengineering Department, Polish Academy of Sciences, Pawinskiego Str., 02-106 Warsaw, Poland
| | - Amit Sangwan
- Department of Electrical Engineering, Northeastern University, Boston, MA 02115, USA; (A.S.); (J.M.J.)
| | - Justin Diehl
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14228, USA; (N.M.); (M.L.); (J.D.); (A.B.); (S.S.); (J.A.); (Y.B.); (E.K.S.)
| | - Anna Balcerak
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14228, USA; (N.M.); (M.L.); (J.D.); (A.B.); (S.S.); (J.A.); (Y.B.); (E.K.S.)
| | - Sneha Sudhakar
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14228, USA; (N.M.); (M.L.); (J.D.); (A.B.); (S.S.); (J.A.); (Y.B.); (E.K.S.)
| | - Justyna Augustyniak
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14228, USA; (N.M.); (M.L.); (J.D.); (A.B.); (S.S.); (J.A.); (Y.B.); (E.K.S.)
- Mossakowski Medical Research Center, Stem Cell Bioengineering Department, Polish Academy of Sciences, Pawinskiego Str., 02-106 Warsaw, Poland
| | - Josep M. Jornet
- Department of Electrical Engineering, Northeastern University, Boston, MA 02115, USA; (A.S.); (J.M.J.)
| | - Yongho Bae
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14228, USA; (N.M.); (M.L.); (J.D.); (A.B.); (S.S.); (J.A.); (Y.B.); (E.K.S.)
| | - Ewa K. Stachowiak
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14228, USA; (N.M.); (M.L.); (J.D.); (A.B.); (S.S.); (J.A.); (Y.B.); (E.K.S.)
| | - Anirban Dutta
- Department of Biomedical Engineering, University at Buffalo, Buffalo, NY 14228, USA; (S.D.); (A.D.)
- Institute of Metabolism and Systems Research, Birmingham Research Park, Birmingham B15 2SQ, UK
| | - Michal K. Stachowiak
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14228, USA; (N.M.); (M.L.); (J.D.); (A.B.); (S.S.); (J.A.); (Y.B.); (E.K.S.)
| |
Collapse
|
2
|
Zhang W, Luo P, Liu X, Cheng R, Zhang S, Qian X, Liu F. Roles of Fibroblast Growth Factors in the Axon Guidance. Int J Mol Sci 2023; 24:10292. [PMID: 37373438 DOI: 10.3390/ijms241210292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/12/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Fibroblast growth factors (FGFs) have been widely studied by virtue of their ability to regulate many essential cellular activities, including proliferation, survival, migration, differentiation and metabolism. Recently, these molecules have emerged as the key components in forming the intricate connections within the nervous system. FGF and FGF receptor (FGFR) signaling pathways play important roles in axon guidance as axons navigate toward their synaptic targets. This review offers a current account of axonal navigation functions performed by FGFs, which operate as chemoattractants and/or chemorepellents in different circumstances. Meanwhile, detailed mechanisms behind the axon guidance process are elaborated, which are related to intracellular signaling integration and cytoskeleton dynamics.
Collapse
Affiliation(s)
- Weiyun Zhang
- Queen Mary School, Medical College, Nanchang University, Nanchang 330000, China
- Medical Experimental Teaching Center, School of Basic Medical Sciences, Nanchang University, Nanchang 330031, China
| | - Peiyi Luo
- Queen Mary School, Medical College, Nanchang University, Nanchang 330000, China
| | - Xiaohan Liu
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Ruoxi Cheng
- Queen Mary School, Medical College, Nanchang University, Nanchang 330000, China
| | - Shuxian Zhang
- Queen Mary School, Medical College, Nanchang University, Nanchang 330000, China
| | - Xiao Qian
- Queen Mary School, Medical College, Nanchang University, Nanchang 330000, China
| | - Fang Liu
- Department of Cell Biology, School of Basic Medical Sciences, Nanchang University, Nanchang 330031, China
| |
Collapse
|
3
|
Chronic Voluntary Alcohol Consumption Alters Promoter Methylation and Expression of Fgf-2 and Fgfr1. Int J Mol Sci 2023; 24:ijms24043336. [PMID: 36834747 PMCID: PMC9963845 DOI: 10.3390/ijms24043336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/02/2023] [Accepted: 02/04/2023] [Indexed: 02/11/2023] Open
Abstract
Alcohol abuse accounts for 3.3 million deaths annually, rendering it a global health issue. Recently, fibroblast growth factor 2 (FGF-2) and its target, fibroblast growth factor receptor 1 (FGFR1), were discovered to positively regulate alcohol-drinking behaviors in mice. We tested whether alcohol intake and withdrawal alter DNA methylation of Fgf-2 and Fgfr1 and if there is a correlation regarding mRNA expression of these genes. Blood and brain tissues of mice receiving alcohol intermittently over a six-week period were analyzed using direct bisulfite sequencing and qRT-PCR analysis. Assessment of Fgf-2 and Fgfr1 promoter methylation revealed changes in the methylation of cytosines in the alcohol group compared with the control group. Moreover, we showed that the altered cytosines coincided with binding motives of several transcription factors. We also found that Fgf-2 and Fgfr1 gene expression was significantly decreased in alcohol-receiving mice compared with control littermates, and that this effect was specifically detected in the dorsomedial striatum, a brain region involved in the circuitry of the reward system. Overall, our data showed alcohol-induced alterations in both mRNA expression and methylation pattern of Fgf-2 and Fgfr1. Furthermore, these alterations showed a reward system regional specificity, therefore, resembling potential targets for future pharmacological interventions.
Collapse
|
4
|
Pan S, Yang PH, DeFreitas D, Ramagiri S, Bayguinov PO, Hacker CD, Snyder AZ, Wilborn J, Huang H, Koller GM, Raval DK, Halupnik GL, Sviben S, Achilefu S, Tang R, Haller G, Quirk JD, Fitzpatrick JAJ, Esakky P, Strahle JM. Gold nanoparticle-enhanced X-ray microtomography of the rodent reveals region-specific cerebrospinal fluid circulation in the brain. Nat Commun 2023; 14:453. [PMID: 36707519 PMCID: PMC9883388 DOI: 10.1038/s41467-023-36083-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 01/12/2023] [Indexed: 01/28/2023] Open
Abstract
Cerebrospinal fluid (CSF) is essential for the development and function of the central nervous system (CNS). However, the brain and its interstitium have largely been thought of as a single entity through which CSF circulates, and it is not known whether specific cell populations within the CNS preferentially interact with the CSF. Here, we develop a technique for CSF tracking, gold nanoparticle-enhanced X-ray microtomography, to achieve micrometer-scale resolution visualization of CSF circulation patterns during development. Using this method and subsequent histological analysis in rodents, we identify previously uncharacterized CSF pathways from the subarachnoid space (particularly the basal cisterns) that mediate CSF-parenchymal interactions involving 24 functional-anatomic cell groupings in the brain and spinal cord. CSF distribution to these areas is largely restricted to early development and is altered in posthemorrhagic hydrocephalus. Our study also presents particle size-dependent CSF circulation patterns through the CNS including interaction between neurons and small CSF tracers, but not large CSF tracers. These findings have implications for understanding the biological basis of normal brain development and the pathogenesis of a broad range of disease states, including hydrocephalus.
Collapse
Affiliation(s)
- Shelei Pan
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Peter H Yang
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Dakota DeFreitas
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Sruthi Ramagiri
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Peter O Bayguinov
- Washington University Center for Cellular Imaging, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Carl D Hacker
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Abraham Z Snyder
- Department of Radiology, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- Department of Neurology, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Jackson Wilborn
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Hengbo Huang
- Department of Radiology, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Gretchen M Koller
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Dhvanii K Raval
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Grace L Halupnik
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Sanja Sviben
- Washington University Center for Cellular Imaging, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Samuel Achilefu
- Department of Biomedical Engineering, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Rui Tang
- Department of Radiology, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Gabriel Haller
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- Department of Neurology, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- Department of Genetics, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - James D Quirk
- Department of Radiology, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - James A J Fitzpatrick
- Washington University Center for Cellular Imaging, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- Department of Neuroscience, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- Department of Cell Biology and Physiology, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Prabagaran Esakky
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Jennifer M Strahle
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA.
- Department of Orthopedic Surgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA.
- Department of Pediatrics, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA.
| |
Collapse
|
5
|
Huang Q, Liu B, Wu W. Biomaterial-Based bFGF Delivery for Nerve Repair. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:8003821. [PMID: 37077657 PMCID: PMC10110389 DOI: 10.1155/2023/8003821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 04/21/2023]
Abstract
Diseases in the nervous system are common in the human body. People have to suffer a great burden due to huge economic costs and poor prognosis of the diseases. Many treatment modalities are now available that can make recovery better. Managing nutritional factors is also helpful for such diseases. The basic fibroblast growth factor (bFGF) is one of the major nutritional factors, which plays a crucial role in organogenesis and tissue homeostasis. It plays a role in cell proliferation, migration, and differentiation, thereby regulating angiogenesis and wound healing and repair of the muscle, bone, and nerve. The study on how to improve the stability of bFGF to increase the treatment effect for different diseases has garnered tremendous attention. Biomaterials are the popular methods to improve the stability of bFGF because they are safe for the living body as they are biocompatible. Biomaterials can be loaded with bFGF and delivered locally to achieve the goal of sustained bFGF release. In the present review, we report different types of biomaterials that are used for bFGF delivery for nerve repair and briefly report how the introduced bFGF can function in the nervous system. We aim to provide summative guidance for future studies about nerve injury using bFGF.
Collapse
Affiliation(s)
- Qinying Huang
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, China
| | - Bo Liu
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, China
| | - Wencan Wu
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, China
| |
Collapse
|
6
|
Prakash N. Developmental pathways linked to the vulnerability of adult midbrain dopaminergic neurons to neurodegeneration. Front Mol Neurosci 2022; 15:1071731. [PMID: 36618829 PMCID: PMC9815185 DOI: 10.3389/fnmol.2022.1071731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
The degeneration of dopaminergic and other neurons in the aging brain is considered a process starting well beyond the infantile and juvenile period. In contrast to other dopamine-associated neuropsychiatric disorders, such as schizophrenia and drug addiction, typically diagnosed during adolescence or young adulthood and, thus, thought to be rooted in the developing brain, Parkinson's Disease (PD) is rarely viewed as such. However, evidences have accumulated suggesting that several factors might contribute to an increased vulnerability to death of the dopaminergic neurons at an already very early (developmental) phase in life. Despite the remarkable ability of the brain to compensate such dopamine deficits, the early loss or dysfunction of these neurons might predispose an individual to suffer from PD because the critical threshold of dopamine function will be reached much earlier in life, even if the time-course and strength of naturally occurring and age-dependent dopaminergic cell death is not markedly altered in this individual. Several signaling and transcriptional pathways required for the proper embryonic development of the midbrain dopaminergic neurons, which are the most affected in PD, either continue to be active in the adult mammalian midbrain or are reactivated at the transition to adulthood and under neurotoxic conditions. The persistent activity of these pathways often has neuroprotective functions in adult midbrain dopaminergic neurons, whereas the reactivation of silenced pathways under pathological conditions can promote the survival and even regeneration of these neurons in the lesioned or aging brain. This article summarizes our current knowledge about signaling and transcription factors involved in midbrain dopaminergic neuron development, whose reduced gene dosage or signaling activity are implicated in a lower survival rate of these neurons in the postnatal or aging brain. It also discusses the evidences supporting the neuroprotection of the midbrain dopaminergic system after the external supply or ectopic expression of some of these secreted and nuclear factors in the adult and aging brain. Altogether, the timely monitoring and/or correction of these signaling and transcriptional pathways might be a promising approach to a much earlier diagnosis and/or prevention of PD.
Collapse
|
7
|
Du X, McManus DP, Fogarty CE, Jones MK, You H. Schistosoma mansoni Fibroblast Growth Factor Receptor A Orchestrates Multiple Functions in Schistosome Biology and in the Host-Parasite Interplay. Front Immunol 2022; 13:868077. [PMID: 35812433 PMCID: PMC9257043 DOI: 10.3389/fimmu.2022.868077] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 05/26/2022] [Indexed: 12/02/2022] Open
Abstract
Stem cells play significant roles in driving the complex life cycle of Schistosoma mansoni. Fibroblast growth factor (FGF) receptor A (SmFGFRA) is essential for maintaining the integrity of schistosome stem cells. Using immunolocalization, we demonstrated that SmFGFRA was distributed abundantly in germinal/stem cells of different S. mansoni life stages including eggs, miracidia, cercariae, schistosomula and adult worms. Indeed, SmFGFRA was also localized amply in embryonic cells and in the perinuclear region of immature eggs; von Lichtenberg's layer and the neural mass of mature eggs; the ciliated surface and neural mass of miracidia; the tegument cytosol of cercariae, schistosomula and adult worms; and was present in abundance in the testis and vitellaria of adult worms of S. mansoni. The distribution pattern of SmFGFRA illustrates the importance of this molecule in maintaining stem cells, development of the nervous and reproductive system of schistosomes, and in the host-parasite interplay. We showed SmFGFRA can bind human FGFs, activating the mitogen activated protein kinase (MAPK) pathway of adult worms in vitro. Inhibition of FGF signaling by the specific tyrosine kinase inhibitor BIBF 1120 significantly reduced egg hatching ability and affected the behavior of miracidia hatched from the treated eggs, emphasizing the importance of FGF signaling in driving the life cycle of S. mansoni. Our findings provide increased understanding of the complex schistosome life cycle and host-parasite interactions, indicating components of the FGF signaling pathway may represent promising targets for developing new interventions against schistosomiasis.
Collapse
Affiliation(s)
- Xiaofeng Du
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Donald P. McManus
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Conor E. Fogarty
- Genecology Research Centre, University of the Sunshine Coast, Brisbane, QLD, Australia
| | - Malcolm K. Jones
- School of Veterinary Science, The University of Queensland, Gatton, QLD, Australia
| | - Hong You
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| |
Collapse
|
8
|
Chang GQ, Yasmin N, Collier AD, Karatayev O, Khalizova N, Onoichenco A, Fam M, Albeg AS, Campbell S, Leibowitz SF. Fibroblast growth factor 2: Role in prenatal alcohol-induced stimulation of hypothalamic peptide neurons. Prog Neuropsychopharmacol Biol Psychiatry 2022; 116:110536. [PMID: 35176416 PMCID: PMC8920779 DOI: 10.1016/j.pnpbp.2022.110536] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 02/08/2022] [Accepted: 02/10/2022] [Indexed: 12/13/2022]
Abstract
Prenatal alcohol exposure (PAE) increases alcohol consumption and risk for alcohol use disorder. This phenomenon in rodents is suggested to involve a stimulatory effect of PAE, in female more than male offspring, on neurogenesis and density of neurons expressing neuropeptides in lateral hypothalamus (LH), including melanin-concentrating hormone (MCH), known to promote alcohol intake. With evidence suggesting a role for fibroblast growth factor 2 (FGF2) and its receptor FGFR1 in stimulating neurogenesis and alcohol drinking, we investigated here whether the FGF2-FGFR1 system is involved in the PAE-induced increase in MCH neurons, in postnatal offspring of pregnant rats given ethanol orally (embryonic day 10-15) at a low-moderate (2 g/kg/day) or high (5 g/kg/day) dose. Our results demonstrate that PAE at the low-moderate but not high dose stimulates FGF2 and FGFR1 gene expression and increases the density of MCH neurons co-expressing FGF2, only in females, but FGFR1 in both sexes. PAE induces this effect in the dorsal but not ventral area of the LH. Further analysis of FGF2 and FGFR1 transcripts within individual MCH neurons reveals an intracellular, sex-dependent effect, with PAE increasing FGF2 transcripts positively related to FGFR1 in the nucleus as well as cytoplasm of females but transcripts only in the cytoplasm of males. Peripheral injection of FGF2 itself (80 μg/kg, s.c.) in pregnant rats mimics these effects of PAE. Together, these results support the involvement of the FGF2-FGFR1 system in mediating the PAE-induced, sex dependent increase in density of MCH neurons, possibly contributing to increased alcohol consumption in the offspring.
Collapse
Affiliation(s)
- Guo-Qing Chang
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065, United States of America
| | - Nushrat Yasmin
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065, United States of America
| | - Adam D Collier
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065, United States of America
| | - Olga Karatayev
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065, United States of America
| | - Nailya Khalizova
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065, United States of America
| | - Amanda Onoichenco
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065, United States of America
| | - Milisia Fam
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065, United States of America
| | - Avi S Albeg
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065, United States of America
| | - Samantha Campbell
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065, United States of America
| | - Sarah F Leibowitz
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065, United States of America.
| |
Collapse
|
9
|
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: 9] [Impact Index Per Article: 4.5] [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
|
10
|
Shafi O, Siddiqui G. Tracing the origins of glioblastoma by investigating the role of gliogenic and related neurogenic genes/signaling pathways in GBM development: a systematic review. World J Surg Oncol 2022; 20:146. [PMID: 35538578 PMCID: PMC9087910 DOI: 10.1186/s12957-022-02602-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 04/15/2022] [Indexed: 02/16/2023] Open
Abstract
Background Glioblastoma is one of the most aggressive tumors. The etiology and the factors determining its onset are not yet entirely known. This study investigates the origins of GBM, and for this purpose, it focuses primarily on developmental gliogenic processes. It also focuses on the impact of the related neurogenic developmental processes in glioblastoma oncogenesis. It also addresses why glial cells are at more risk of tumor development compared to neurons. Methods Databases including PubMed, MEDLINE, and Google Scholar were searched for published articles without any date restrictions, involving glioblastoma, gliogenesis, neurogenesis, stemness, neural stem cells, gliogenic signaling and pathways, neurogenic signaling and pathways, and astrocytogenic genes. Results The origin of GBM is dependent on dysregulation in multiple genes and pathways that accumulatively converge the cells towards oncogenesis. There are multiple layers of steps in glioblastoma oncogenesis including the failure of cell fate-specific genes to keep the cells differentiated in their specific cell types such as p300, BMP, HOPX, and NRSF/REST. There are genes and signaling pathways that are involved in differentiation and also contribute to GBM such as FGFR3, JAK-STAT, and hey1. The genes that contribute to differentiation processes but also contribute to stemness in GBM include notch, Sox9, Sox4, c-myc gene overrides p300, and then GFAP, leading to upregulation of nestin, SHH, NF-κB, and others. GBM mutations pathologically impact the cell circuitry such as the interaction between Sox2 and JAK-STAT pathway, resulting in GBM development and progression. Conclusion Glioblastoma originates when the gene expression of key gliogenic genes and signaling pathways become dysregulated. This study identifies key gliogenic genes having the ability to control oncogenesis in glioblastoma cells, including p300, BMP, PAX6, HOPX, NRSF/REST, LIF, and TGF beta. It also identifies key neurogenic genes having the ability to control oncogenesis including PAX6, neurogenins including Ngn1, NeuroD1, NeuroD4, Numb, NKX6-1 Ebf, Myt1, and ASCL1. This study also postulates how aging contributes to the onset of glioblastoma by dysregulating the gene expression of NF-κB, REST/NRSF, ERK, AKT, EGFR, and others.
Collapse
Affiliation(s)
- Ovais Shafi
- Sindh Medical College - Jinnah Sindh Medical University / Dow University of Health Sciences, Karachi, Pakistan.
| | - Ghazia Siddiqui
- Sindh Medical College - Jinnah Sindh Medical University / Dow University of Health Sciences, Karachi, Pakistan
| |
Collapse
|
11
|
Bono BS, Koziel Ly NK, Miller PA, Williams-Ikhenoba J, Dumiaty Y, Chee MJ. Spatial distribution of beta-klotho mRNA in the mouse hypothalamus, hippocampal region, subiculum, and amygdala. J Comp Neurol 2022; 530:1634-1657. [PMID: 35143049 DOI: 10.1002/cne.25306] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 01/05/2022] [Accepted: 01/20/2022] [Indexed: 11/10/2022]
Abstract
Beta-klotho (KLB) is a co-receptor required for endocrine fibroblast growth factor (FGF) 15/19 and FGF21 signaling in the brain. Klb is prominent within the hypothalamus, which is consistent with its metabolic functions, but diverse roles for Klb are now emerging. Central Klb expression is low but discrete and may govern FGF-targeted sites. However, given its low expression, it is unclear if Klb mRNA is more widespread. We performed in situ hybridization to label Klb mRNA to generate spatial maps capturing the distribution and level of Klb within the mouse hypothalamus, hippocampal region, subiculum, and amygdala. Semi-quantitative analysis revealed that Klb-labeled cells may express low, medium, or high levels of Klb mRNA. Hypothalamic Klb hybridization was heterogeneous and varied rostrocaudally within the same region. Most Klb-labeled cells were found in the lateral hypothalamic zone, but the periventricular hypothalamic region, including the suprachiasmatic nucleus, contained the greatest proportion of cells expressing medium or high Klb levels. We also found heterogeneous Klb hybridization in the amygdala and subiculum, where Klb was especially distinct within the central amygdalar nucleus and ventral subiculum, respectively. By contrast, Klb-labeled cells in the hippocampal region only expressed low levels of Klb and were typically found in the pyramidal layer of Ammon's horn or dentate gyrus. The Klb-labeled regions identified in this study are consistent with reported roles of Klb in metabolism, taste preference, and neuroprotection. However, additional identified sites, including within the hypothalamus and amygdala, may suggest novel roles for FGF15/19 or FGF21 signaling. The central expression of beta-klotho (Klb) is essential for the physiological actions of endocrine fibroblast growth factors. Klb mRNA was widely expressed throughout the hypothalamus, hippocampus, and amygdala. However, the level of Klb expression varied between cells and contributed to a distinctive pattern of distribution within each brain structure. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Bianca S Bono
- Department of Neuroscience, Carleton University, Ottawa, ON, K1S 5B6, Canada
| | - Nikita K Koziel Ly
- Department of Neuroscience, Carleton University, Ottawa, ON, K1S 5B6, Canada
| | - Persephone A Miller
- Department of Neuroscience, Carleton University, Ottawa, ON, K1S 5B6, Canada
| | | | - Yasmina Dumiaty
- Department of Neuroscience, Carleton University, Ottawa, ON, K1S 5B6, Canada
| | - Melissa J Chee
- Department of Neuroscience, Carleton University, Ottawa, ON, K1S 5B6, Canada
| |
Collapse
|
12
|
Wang Y, Zhang Y, Shi Z, Di T, Yu W, Chen L. Exposure of male mice to perfluorooctanoic acid induces anxiety-like behaviors by increasing corticotropin-releasing factor in the basolateral amygdala complex. CHEMOSPHERE 2022; 287:132170. [PMID: 34826932 DOI: 10.1016/j.chemosphere.2021.132170] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 08/16/2021] [Accepted: 09/02/2021] [Indexed: 06/13/2023]
Abstract
Perfluorooctanoic acid (PFOA), a hazardous environmental pollutant, has been found to enhance hepatic synthesis of fibroblast growth factor 21 (FGF21). FGF21 can enter the brain and increase the expression of corticotropin-releasing factor (CRF) in the paraventricular nucleus (PVN). In this study, adult male mice were orally administered PFOA to evaluate how it regulates emotion. Exposure of mice to PFOA (1 mg kg-1 bw) for 10 consecutive days (PFOA-mice) caused anxiety-like behaviors and a peroxisome proliferator-activated receptor α (PPARα)-dependent increase in hepatic FGF21 synthesis. The levels of CRF expression in not only PVN but also basolateral amygdala complex (BLA) neurons of PFOA-mice were increased via FGF receptor 1 (FGF-R1) activation. However, the microinjection of FGF-R1 or CRF 1 receptor (CRF-R1) antagonist in the BLA rather than the PVN of PFOA-mice could relieve their anxiety-like behaviors. In addition, external capsule-BLA synaptic transmission in PFOA-mice was enhanced by increasing CRF-R1-mediated presynaptic glutamate release, which was corrected by the blockade of PPARα, FGF-R1 and CRF-R1 or the inhibition of PKA. Furthermore, the threshold of frequency-dependent long-term potentiation (LTP) induction was decreased in the BLA of PFOA-mice, which depended on the activation of PPARα, FGF-R1, CRF-R1, PKA and NMDA receptor (NMDAR), whereas long-term depression (LTD) induction was unchanged. Thus, the results indicate that the exposure of male mice to PFOA (1 mg kg-1 bw) enhances CRF expression in BLA neurons by increasing hepatic FGF21 synthesis, which then enhances CRF-R1-mediated presynaptic glutamate release to facilitate NMDAR-dependent BLA-LTP induction, leading to the production of anxiety-like behaviors.
Collapse
Affiliation(s)
- Ya Wang
- Department of Physiology, Nanjing Medical University, Nanjing, 211166, China
| | - Yajie Zhang
- Department of Physiology, Nanjing Medical University, Nanjing, 211166, China
| | - Zhaochun Shi
- Department of Neurology, First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, China
| | - Tingting Di
- Department of Physiology, Nanjing Medical University, Nanjing, 211166, China
| | - Wenfeng Yu
- Key Laboratory of Medical Molecular Biology, Guizhou Medical University, Guiyang, 550004, China.
| | - Ling Chen
- Department of Physiology, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
13
|
Tennakoon A, Katharesan V, Musgrave IF, Koblar SA, Faull RLM, Curtis MA, Johnson IP. Normal aging, motor neurone disease, and Alzheimer's disease are characterized by cortical changes in inflammatory cytokines. J Neurosci Res 2021; 100:653-669. [PMID: 34882833 DOI: 10.1002/jnr.24996] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 10/29/2021] [Accepted: 11/06/2021] [Indexed: 12/12/2022]
Abstract
The role of increased brain inflammation in the development of neurodegenerative diseases is unclear. Here, we have compared cytokine changes in normal aging, motor neurone disease (MND), and Alzheimer's disease (AD). After an initial analysis, six candidate cytokines, interleukin (IL)- 4, 5, 6, 10, macrophage inhibitory protein (MIP)-1α, and fibroblast growth factor (FGF)-2, showing greatest changes were assayed in postmortem frozen human superior frontal gyri (n = 12) of AD patients, aging and young adult controls along with the precentral gyrus (n = 12) of MND patients. Healthy aging was associated with decreased anti-inflammatory IL-10 and FGF-2 levels. AD prefrontal cortex was associated with increased levels of IL-4, IL-5, and FGF-2, with the largest increase seen for FGF-2. Notwithstanding differences in the specific frontal lobe gyrus sampled, MND patients' primary motor cortex (precentral gyrus) was associated with increased levels of IL-5, IL-6, IL-10, and FGF-2 compared to the aging prefrontal cortex (superior frontal gyrus). Immunocytochemistry showed that FGF-2 is expressed in neurons, astrocytes, and microglia in normal aging prefrontal cortex, AD prefrontal cortex, and MND motor cortex. We report that healthy aging and age-related neurodegenerative diseases have different cortical inflammatory signatures that are characterized by increased levels of anti-inflammatory cytokines and call into question the view that increased inflammation underlies the development of age-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Anuradha Tennakoon
- School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - Viythia Katharesan
- School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | | | - Simon Andrea Koblar
- Department of Medical Specialties, Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Richard Lewis Maxwell Faull
- Department of Anatomy and Medical Imaging and the Centre for Brain Research, The University of Auckland, Auckland, New Zealand
| | - Maurice Anthony Curtis
- Department of Anatomy and Medical Imaging and the Centre for Brain Research, The University of Auckland, Auckland, New Zealand
| | - Ian Paul Johnson
- Macquarie Medical School, Macquarie University, Sydney, NSW, Australia
| |
Collapse
|
14
|
Dremencov E, Jezova D, Barak S, Gaburjakova J, Gaburjakova M, Kutna V, Ovsepian SV. Trophic factors as potential therapies for treatment of major mental disorders. Neurosci Lett 2021; 764:136194. [PMID: 34433100 DOI: 10.1016/j.neulet.2021.136194] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 08/02/2021] [Accepted: 08/20/2021] [Indexed: 12/20/2022]
Abstract
Notwithstanding major advances in psychotherapeutics, their efficacy and specificity remain limited. The slow onset of beneficial outcomes and numerous adverse effects of widely used medications remain of chief concern, warranting in-depth studies. The majority of frontline therapies are thought to enhance the endogenous monoaminergic drive, to initiate a cascade of molecular events leading to lasting functional and structural plasticity. They also involve alterations in trophic factor signalling, including brain-derived neurotrophic factor (BDNF), VGF (non-acronymic), vascular endothelial growth factor (VEGF), fibroblast growth factor 2 (FGF2), glial cell-derived neurotrophic factor (GDNF), and others. In several major mental disorders, emerging data suggest protective and restorative effects of trophic factors in preclinical models, when applied on their own. Antidepressant outcomes of VGF and FGF2, for instance, were shown in experimental animals, while BDNF and GDNF prove useful in the treatment of addiction, schizophrenia, and autism spectrum disorders. The main challenge with the effective translation of these and other findings in the clinic is the knowledge gap in action mechanisms with potential risks, as well as the lack of effective platforms for validation under clinical settings. Herein, we review the state-of-the-art and advances in the therapeutic use of trophic factors in several major neuropsychiatric disorders.
Collapse
Affiliation(s)
- Eliyahu Dremencov
- Institute of Molecular Physiology and Genetics, Center of Biosciences, Slovak Academy of Sciences, Bratislava, Slovakia.
| | - Daniela Jezova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Segev Barak
- School of Psychological Sciences and the Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Jana Gaburjakova
- Institute of Molecular Physiology and Genetics, Center of Biosciences, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Marta Gaburjakova
- Institute of Molecular Physiology and Genetics, Center of Biosciences, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Viera Kutna
- Department of Experimental Neurobiology, National Institute of Mental Health, Topolová 748, 250 67 Klecany, Czech Republic
| | - Saak V Ovsepian
- Department of Experimental Neurobiology, National Institute of Mental Health, Topolová 748, 250 67 Klecany, Czech Republic
| |
Collapse
|
15
|
Xia J, Xue X, Liu W, Qi Z, Liu W. The Role of Fgf9 in the Antidepressant Effects of Exercise and Fluoxetine in Chronic Unpredictable Mild Stress Mice. Psychosom Med 2021; 83:795-804. [PMID: 33938506 DOI: 10.1097/psy.0000000000000953] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
OBJECTIVE The neurotrophic hypothesis of depression posits that stress and depression decrease neurotrophic factor expression in brain, whereas antidepressants and exercise can contribute to the blockade of stress effects and produce antidepressant effects. Fibroblast growth factor 9 (FGF9), a member of the fibroblast growth factor (FGF) family, has been reported to be dysregulated in depression. The present study aimed to determine whether and how Fgf9 mediates the antidepressant effects of fluoxetine and exercise in chronic unpredictable mild stress (CUMS) mice. METHODS Male C57BL/6 mice were exposed to CUMS for 7 weeks. From the fourth week, CUMS-exposed mice were subjected to fluoxetine treatment or swimming exercise for 4 weeks. Forced swim test, tail suspension test, and hole-board test were used to assess behaviors of mice. Real-time polymerase chain reaction was used to examine hippocampal messenger RNA levels of Fgf9, Fgf2, FgfR1, FgfR2, and FgfR3. Western blotting was used to examine the protein levels of Fgf9, protein kinase B (Akt), and phosphorylation of Akt at Ser473 in mouse hippocampus. RESULTS Our results demonstrated that CUMS induced depression-like behaviors, which were reversed by fluoxetine treatment and swimming exercise. Moreover, we found that CUMS resulted in a dysregulation of Fgf9, Fgf2, and FgfR2 expression, whereas fluoxetine and swimming restored the FGF expression in CUMS-exposed mice. An analysis of the proteins suggests that the antidepressant effects of fluoxetine and exercise in CUMS-exposed mice were associated with ameliorated Fgf9/Akt signaling. CONCLUSIONS Our findings have demonstrated that swimming exercise mimics the antidepressant effects of fluoxetine by regulating Fgf9 in CUMS-exposed mice, which may offer new mechanism-based therapeutic targets for depression.
Collapse
Affiliation(s)
- Jie Xia
- From the Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education (Xia, Xue, Wenbin Liu, Qi, Weina Liu), College of Physical Education and Health (Xia, Wenbin Liu, Qi, Weina Liu), East China Normal University; and Key Laboratory of Exercise and Health Sciences of Ministry of Education (Xue), Shanghai University of Sport, Shanghai, China
| | | | | | | | | |
Collapse
|
16
|
Klimaschewski L, Claus P. Fibroblast Growth Factor Signalling in the Diseased Nervous System. Mol Neurobiol 2021; 58:3884-3902. [PMID: 33860438 PMCID: PMC8280051 DOI: 10.1007/s12035-021-02367-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 03/19/2021] [Indexed: 12/12/2022]
Abstract
Fibroblast growth factors (FGFs) act as key signalling molecules in brain development, maintenance, and repair. They influence the intricate relationship between myelinating cells and axons as well as the association of astrocytic and microglial processes with neuronal perikarya and synapses. Advances in molecular genetics and imaging techniques have allowed novel insights into FGF signalling in recent years. Conditional mouse mutants have revealed the functional significance of neuronal and glial FGF receptors, not only in tissue protection, axon regeneration, and glial proliferation but also in instant behavioural changes. This review provides a summary of recent findings regarding the role of FGFs and their receptors in the nervous system and in the pathogenesis of major neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Lars Klimaschewski
- Department of Anatomy, Histology and Embryology, Institute of Neuroanatomy, Medical University of Innsbruck, Innsbruck, Austria.
| | - Peter Claus
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Hannover, Germany
- Center for Systems Neuroscience, Hannover, Germany
| |
Collapse
|
17
|
Gascon S, Jann J, Langlois-Blais C, Plourde M, Lavoie C, Faucheux N. Peptides Derived from Growth Factors to Treat Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22116071. [PMID: 34199883 PMCID: PMC8200100 DOI: 10.3390/ijms22116071] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 05/30/2021] [Accepted: 06/01/2021] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disease characterized by progressive neuron losses in memory-related brain structures. The classical features of AD are a dysregulation of the cholinergic system, the accumulation of amyloid plaques, and neurofibrillary tangles. Unfortunately, current treatments are unable to cure or even delay the progression of the disease. Therefore, new therapeutic strategies have emerged, such as the exogenous administration of neurotrophic factors (e.g., NGF and BDNF) that are deficient or dysregulated in AD. However, their low capacity to cross the blood-brain barrier and their exorbitant cost currently limit their use. To overcome these limitations, short peptides mimicking the binding receptor sites of these growth factors have been developed. Such peptides can target selective signaling pathways involved in neuron survival, differentiation, and/or maintenance. This review focuses on growth factors and their derived peptides as potential treatment for AD. It describes (1) the physiological functions of growth factors in the brain, their neuronal signaling pathways, and alteration in AD; (2) the strategies to develop peptides derived from growth factor and their capacity to mimic the role of native proteins; and (3) new advancements and potential in using these molecules as therapeutic treatments for AD, as well as their limitations.
Collapse
Affiliation(s)
- Suzanne Gascon
- Laboratory of Cell-Biomaterial Biohybrid Systems, Department of Chemical and Biotechnological Engineering, 2500 Boulevard Université, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada; (S.G.); (J.J.)
| | - Jessica Jann
- Laboratory of Cell-Biomaterial Biohybrid Systems, Department of Chemical and Biotechnological Engineering, 2500 Boulevard Université, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada; (S.G.); (J.J.)
| | - Chloé Langlois-Blais
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
| | - Mélanie Plourde
- Centre de Recherche sur le Vieillissement, Centre Intégré Universitaire de Santé et Services Sociaux de l’Estrie–Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1G 1B1, Canada;
- Département de Médecine, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Christine Lavoie
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
- Institut de Pharmacologie de Sherbrooke, 3001 12th Avenue, N., Sherbrooke, QC J1H 5N4, Canada
- Correspondence: (C.L.); (N.F.); Tel.: +1-819-821-8000 (ext. 72732) (C.L.); +1-819-821-8000 (ext. 61343) (N.F.)
| | - Nathalie Faucheux
- Laboratory of Cell-Biomaterial Biohybrid Systems, Department of Chemical and Biotechnological Engineering, 2500 Boulevard Université, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada; (S.G.); (J.J.)
- Institut de Pharmacologie de Sherbrooke, 3001 12th Avenue, N., Sherbrooke, QC J1H 5N4, Canada
- Correspondence: (C.L.); (N.F.); Tel.: +1-819-821-8000 (ext. 72732) (C.L.); +1-819-821-8000 (ext. 61343) (N.F.)
| |
Collapse
|
18
|
Alan E, Liman N. The distribution and immunolocalization of fibroblast growth factors (FGFs) in the rat oviduct during early pregnancy and the post-partum period. Anat Histol Embryol 2021; 50:645-657. [PMID: 33772852 DOI: 10.1111/ahe.12672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 02/28/2021] [Accepted: 03/15/2021] [Indexed: 11/29/2022]
Abstract
The mammalian oviduct provides a favourable environment for several reproductive processes, including ovum transport, sperm capacitation, fertilization and pre-implantation embryonic development. This environment is regulated by cyclic ovarian steroids, that is oestrogen, and growth factors. Fibroblast growth factors (FGFs) regulate the differentiation and growth of various cell types in the female genital tract. This study aimed to determine the localization of FGF1, FGF2, FGF receptor 1 (FGFR1) and 2 (FGFR2) in the rat oviduct, by immunohistochemistry, on day 5 of pregnancy and post-partum days 1, 3 and 5, and to demonstrate the possible functions of these proteins during early pregnancy and the post-partum period. On all examination days, cytoplasmic and nuclear FGF1 immunoreactivity was detected in the epithelium lining the infundibulum, ampulla and isthmus of the oviduct. Immunoreactivity was much stronger in the basal bodies of the cilia on the epithelium lining the infundibulum and ampulla. FGF1 immunoreactivity was also detected in stromal cells, myocytes and endothelial cells. Cytoplasmic FGF2 immunoreactivity was observed in the tunica muscularis, vascular myocytes and endothelial cells. While strong cytoplasmic FGF2 immunoreactions were observed in the stromal cells of the lamina propria, the luminal epithelium, some stromal cells and smooth muscle cells displayed a rather weak FGFR1 and FGFR2 immunoreactivity. Immunoreaction intensity did not differ between the periods examined. This study shows that FGF1, FGF2, FGFR1 and FGFR2 are produced by rat oviduct cells during pregnancy and the post-partum period, and reproductive physiology is regulated not only by hormonal mechanisms, but also by growth factors.
Collapse
Affiliation(s)
- Emel Alan
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Erciyes University, Kayseri, Turkey
| | - Narin Liman
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Erciyes University, Kayseri, Turkey
| |
Collapse
|
19
|
Liran M, Rahamim N, Ron D, Barak S. Growth Factors and Alcohol Use Disorder. Cold Spring Harb Perspect Med 2020; 10:cshperspect.a039271. [PMID: 31964648 DOI: 10.1101/cshperspect.a039271] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Neurotrophic growth factors were originally characterized for their support in neuronal differentiation, outgrowth, and survival during development. However, it has been acknowledged that they also play a vital role in the adult brain. Abnormalities in growth factors have been implicated in a variety of neurological and psychiatric disorders, including alcohol use disorder (AUD). This work focuses on the interaction between alcohol and growth factors. We review literature suggesting that several growth factors play a unique role in the regulation of alcohol consumption, and that breakdown in these growth factor systems is linked to the development of AUD. Specifically, we focus on the brain-derived neurotrophic factor (BDNF), glial cell line-derived neurotrophic factor (GDNF), fibroblast growth factor 2 (FGF2), and insulin growth factor 1 (IGF-1). We also review the literature on the potential role of midkine (MDK) and pleiotrophin (PTN) and their receptor, anaplastic lymphoma kinase (ALK), in AUD. We show that alcohol alters the expression of these growth factors or their receptors in brain regions previously implicated in addiction, and that manipulations on these growth factors and their downstream signaling can affect alcohol-drinking behaviors in animal models. We conclude that there is a need for translational and clinical research to assess the therapeutic potential of new pharmacotherapies targeting these systems.
Collapse
Affiliation(s)
- Mirit Liran
- Department of Neurobiology, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Nofar Rahamim
- Sagol School of Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Dorit Ron
- Department of Neurology, University of California, 675 Nelson Rising Lane, San Francisco, California 94143-0663, USA
| | - Segev Barak
- Department of Neurobiology, Tel Aviv University, 69978 Tel Aviv, Israel.,Sagol School of Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel.,School of Psychological Sciences, Tel Aviv University, 69978 Tel Aviv, Israel
| |
Collapse
|
20
|
Jiang G, Xiao G, Luo C, Tang Z, Teng Z, Peng X. Correlation Between SNPs at the 3'UTR of the FGF2 Gene and Their Interaction with Environmental Factors in Han Chinese Diabetic Peripheral Neuropathy Patients. J Mol Neurosci 2020; 71:203-214. [PMID: 32613556 DOI: 10.1007/s12031-020-01641-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Accepted: 06/19/2020] [Indexed: 12/12/2022]
Abstract
FGF2 is a neurotrophic factor that can act as a key regulatory molecule of neuroprotection, neurogenesis, and angiogenesis in various injuries. To explore the genetic background of the FGF2 gene on DPN development, this study analyzed the correlation between SNPs in the 3'UTR of the FGF2 gene and their interaction with environmental factors in DPN patients of Han Chinese nationality. Sanger sequencing was used to analyze the FGF2 genotypes at the rs1048201, rs3804158, rs41348645, rs6854081, rs3747676, rs7683093, rs1476215, and rs1476217 loci in 150 DPN patients, 150 NDPN patients, and 150 healthy control patients. Plasma FGF2 levels were measured in all subjects by using ELISAs. Subjects carrying the T allele at the rs1048201 locus in the FGF2 gene had a significantly lower risk of developing DPN compared with subjects carrying the C allele (OR = 0.43, 95% CI = 0.33-0.56, p < 0.01). Subjects with the G genotype at the rs6854081 locus had an exceptionally higher risk of developing DPN than subjects with the T allele (OR = 1.66, 95% CI = 1.39-1.89, p < 0.01). Individuals harboring the G allele at the rs7683093 locus had a markedly higher risk of DPN than patients with the C allele (OR = 1.63, 95% CI = 1.36-1.87, p < 0.01). Finally, individuals having the A genotype at the rs1476215 locus had a significantly higher risk of DPN than individuals carrying the T allele (OR = 1.82, 95% CI = 1.53-2.02, p < 0.01). There was an interaction between age and alcohol consumption and the SNP rs7683093. SNPs at rs1048201, rs6854081, rs7683093, and rs1476215 in the FGF2 3'UTR were strongly associated with plasma levels of FGF2 (p < 0.05). SNPs at the rs1048201, rs6854081, rs7683093, and rs1476215 loci in the FGF2 gene were significantly associated with the risk of DPN. A possible mechanism is that these SNPs affect the expression level of FGF2 by interrupting the binding of microRNAs to target sites in the 3'UTR.
Collapse
Affiliation(s)
- Guangyuan Jiang
- Department of Neurosurgery, The Chongqing City Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Gang Xiao
- Department of Neurosurgery, The Chongqing City Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Chao Luo
- Department of Neurosurgery, The Chongqing City Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Zhaohua Tang
- Departmen of neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhipeng Teng
- Department of Neurosurgery, The Chongqing City Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Xing Peng
- Department of Neurosurgery, The Chongqing City Hospital of Traditional Chinese Medicine, Chongqing, China.
| |
Collapse
|
21
|
Even-Chen O, Barak S. Inhibition of FGF Receptor-1 Suppresses Alcohol Consumption: Role of PI3 Kinase Signaling in Dorsomedial Striatum. J Neurosci 2019; 39:7947-7957. [PMID: 31375540 PMCID: PMC6774404 DOI: 10.1523/jneurosci.0805-19.2019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 07/20/2019] [Accepted: 07/25/2019] [Indexed: 12/11/2022] Open
Abstract
Excessive alcohol intake leads to mesostriatal neuroadaptations, and to addiction phenotypes. We recently found in rodents that alcohol increases fibroblast growth factor 2 (FGF2) expression in the dorsomedial striatum (DMS), which promotes alcohol consumption. Here, we show that systemic or intra-DMS blockade of the FGF2 receptor, FGF receptor-1 (FGFR1), suppresses alcohol consumption, and that the effects of FGF2-FGFR1 on alcohol drinking are mediated via the phosphoinositide 3 kinase (PI3K) signaling pathway. Specifically, we found that sub-chronic alcohol treatment (7 d × 2.5 g/kg, i.p.) increased Fgfr1 mRNA expression in the dorsal hippocampus and dorsal striatum. However, prolonged and excessive voluntary alcohol consumption in a two-bottle choice procedure increased Fgfr1 expression selectively in DMS. Importantly, systemic administration of the FGFR1 inhibitor PD173074 to mice, as well as its infusion into the DMS of rats, decreased alcohol consumption and preference, with no effects on natural reward consumption. Finally, inhibition of the PI3K, but not of the mitogen-activated protein kinase (MAPK) signaling pathway, blocked the effects of FGF2 on alcohol intake and preference. Our results suggest that activation of FGFR1 by FGF2 in the DMS leads to activation of the PI3K signaling pathway, which promotes excessive alcohol consumption, and that inhibition of FGFR1 may provide a novel therapeutic target for alcohol use disorder.SIGNIFICANCE STATEMENT Long-term alcohol consumption causes neuroadaptations in the mesostriatal reward system, leading to addiction-related behaviors. We recently showed that alcohol upregulates the expression of fibroblast growth factor 2 (FGF2) in dorsomedial striatum (DMS) or rats and mice, and in turn, FGF2 increases alcohol consumption. Here, we show that long-term alcohol intake also increases the expression of the FGF2 receptor, FGFR1 in the DMS. Importantly, inhibition of FGFR1 activity by a selective receptor antagonist reduces alcohol drinking, when given systemically or directly into the DMS. We further show that the effects of FGF2-FGFR1 on alcohol drinking are mediated via activation of the PI3K intracellular signaling pathway, providing an insight on the mechanism for this effect.
Collapse
Affiliation(s)
| | - Segev Barak
- School of Psychological Sciences, and
- Sagol School of Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel
| |
Collapse
|
22
|
Jimenez-Pascual A, Siebzehnrubl FA. Fibroblast Growth Factor Receptor Functions in Glioblastoma. Cells 2019; 8:E715. [PMID: 31337028 PMCID: PMC6678715 DOI: 10.3390/cells8070715] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 07/11/2019] [Accepted: 07/11/2019] [Indexed: 12/20/2022] Open
Abstract
Glioblastoma is the most lethal brain cancer in adults, with no known cure. This cancer is characterized by a pronounced genetic heterogeneity, but aberrant activation of receptor tyrosine kinase signaling is among the most frequent molecular alterations in glioblastoma. Somatic mutations of fibroblast growth factor receptors (FGFRs) are rare in these cancers, but many studies have documented that signaling through FGFRs impacts glioblastoma progression and patient survival. Small-molecule inhibitors of FGFR tyrosine kinases are currently being trialed, underlining the therapeutic potential of blocking this signaling pathway. Nevertheless, a comprehensive overview of the state of the art of the literature on FGFRs in glioblastoma is lacking. Here, we review the evidence for the biological functions of FGFRs in glioblastoma, as well as pharmacological approaches to targeting these receptors.
Collapse
MESH Headings
- Brain Neoplasms/metabolism
- Disease Progression
- Glioblastoma/metabolism
- Humans
- Receptor, Fibroblast Growth Factor, Type 1/chemistry
- Receptor, Fibroblast Growth Factor, Type 1/physiology
- Receptor, Fibroblast Growth Factor, Type 2/chemistry
- Receptor, Fibroblast Growth Factor, Type 2/physiology
- Receptor, Fibroblast Growth Factor, Type 3/chemistry
- Receptor, Fibroblast Growth Factor, Type 3/physiology
- Receptor, Fibroblast Growth Factor, Type 4/chemistry
- Receptor, Fibroblast Growth Factor, Type 4/physiology
Collapse
Affiliation(s)
- Ana Jimenez-Pascual
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Cardiff CF24 4HQ, UK
| | - Florian A Siebzehnrubl
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Cardiff CF24 4HQ, UK.
| |
Collapse
|
23
|
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
|
24
|
Simard S, Shail P, MacGregor J, El Sayed M, Duman RS, Vaccarino FM, Salmaso N. Fibroblast growth factor 2 is necessary for the antidepressant effects of fluoxetine. PLoS One 2018; 13:e0204980. [PMID: 30273396 PMCID: PMC6166983 DOI: 10.1371/journal.pone.0204980] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 09/18/2018] [Indexed: 01/01/2023] Open
Abstract
Previous research has shown that fibroblast growth factor 2 protein (FGF2) can act as an anxiolytic and anti-depressive agent in rodents. Levels of hippocampal FGF2 and FGF2 receptors are decreased in post-mortem brains of individuals with mood disorders. No changes in FGF2 were noted in the post-mortem brains of individuals with mood disorders that were successfully treated with anti-depressant medication prior to death. Mutations in the FGF2 gene in humans have been shown to predict non-responsiveness to the therapeutic effects of selective serotonin reuptake inhibitors (SSRIs). These findings suggest that FGF2 may potentially be a target of and/or required for the therapeutic effects of antidepressant medications. To test this, we employed a rodent model of depressive behaviour, chronic variable stress (CVS) in conjunction with antidepressant treatment (fluoxetine) in wild-type (WT) and FGF2 knockout mice (FGF2KO) and examined depressive and anxiety behaviors. Results showed that fluoxetine reversed the effects of CVS on depressive and anxiety behaviours in wild-type mice only, suggesting that the FGF2 gene is indeed necessary for the therapeutic effects of fluoxetine. Interestingly, CVS decreased hippocampal FGF2 levels and fluoxetine partially reversed this effect. Because FGF2 has been previously shown to modify HPA activity through hippocampal glucocorticoid receptors (GR), we examined levels of glucocorticoid receptors and found a decrease in GR in response to CVS, with a further decrease in FGF2KO. No effect of fluoxetine on GR was observed in either WT or FGF2KO mice. This suggests that further changes in glucocorticoid receptors are not necessary for the anti-depressant effects of fluoxetine in WT mice, although decreased glucocorticoid receptors in response to FGF2 deletion may preclude the therapeutic actions of fluoxetine in FGF2KO. Whether astroglia, astroglial functions, or HPA changes are the downstream target of FGF2-mediated changes induced by fluoxetine remains to be determined, however, the current study reaffirms the potential of FGF2 as a novel therapeutic target in the treatment of depression and anxiety disorders.
Collapse
Affiliation(s)
- Stephanie Simard
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Pragya Shail
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Jessica MacGregor
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Maha El Sayed
- Department of Psychiatry, Yale University, New Haven, Connecticut, United States of America
| | - Ronald S Duman
- Department of Psychiatry, Yale University, New Haven, Connecticut, United States of America
| | - Flora M Vaccarino
- Child Study Center, Yale University, New Haven, Connecticut, United States of America
| | - Natalina Salmaso
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada.,Child Study Center, Yale University, New Haven, Connecticut, United States of America
| |
Collapse
|
25
|
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
|
26
|
Li HJ, Sun ZL, Yang XT, Zhu L, Feng DF. Exploring Optic Nerve Axon Regeneration. Curr Neuropharmacol 2018; 15:861-873. [PMID: 28029073 PMCID: PMC5652030 DOI: 10.2174/1570159x14666161227150250] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Revised: 12/14/2016] [Accepted: 12/22/2016] [Indexed: 12/13/2022] Open
Abstract
Background: Traumatic optic nerve injury is a leading cause of irreversible blindness across the world and causes progressive visual impairment attributed to the dysfunction and death of retinal ganglion cells (RGCs). To date, neither pharmacological nor surgical interventions are sufficient to halt or reverse the progress of visual loss. Axon regeneration is critical for functional recovery of vision following optic nerve injury. After optic nerve injury, RGC axons usually fail to regrow and die, leading to the death of the RGCs and subsequently inducing the functional loss of vision. However, the detailed molecular mechanisms underlying axon regeneration after optic nerve injury remain poorly understood. Methods: Research content related to the detailed molecular mechanisms underlying axon regeneration after optic nerve injury have been reviewed. Results: The present review provides an overview of regarding potential strategies for axonal regeneration of RGCs and optic nerve repair, focusing on the role of cytokines and their downstream signaling pathways involved in intrinsic growth program and the inhibitory environment together with axon guidance cues for correct axon guidance. A more complete understanding of the factors limiting axonal regeneration will provide a rational basis, which contributes to develop improved treatments for optic nerve regeneration. These findings are encouraging and open the possibility that clinically meaningful regeneration may become achievable in the future. Conclusion: Combination of treatments towards overcoming growth-inhibitory molecules and enhancing intrinsic growth capacity combined with correct guidance using axon guidance cues is crucial for developing promising therapies to promote axon regeneration and functional recovery after ON injury.
Collapse
Affiliation(s)
- Hong-Jiang Li
- Department of Neurosurgery, No.9 People's Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 201999, China
| | - Zhao-Liang Sun
- Department of Neurosurgery, No.9 People's Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 201999, China
| | - Xi-Tao Yang
- Department of Neurosurgery, No.9 People's Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 201999, China
| | - Liang Zhu
- Department of Neurosurgery, No.9 People's Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 201999, China
| | - Dong-Fu Feng
- Department of Neurosurgery, No.9 People's Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 201999, China
| |
Collapse
|
27
|
Vidovic D, Davila RA, Gronostajski RM, Harvey TJ, Piper M. Transcriptional regulation of ependymal cell maturation within the postnatal brain. Neural Dev 2018; 13:2. [PMID: 29452604 PMCID: PMC5816376 DOI: 10.1186/s13064-018-0099-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 02/12/2018] [Indexed: 12/11/2022] Open
Abstract
Background Radial glial stem cells within the developing nervous system generate a variety of post-mitotic cells, including neurons and glial cells, as well as the specialised multi-ciliated cells that line the walls of the ventricular system, the ependymal cells. Ependymal cells separate the brain parenchyma from the cerebrospinal fluid and mediate osmotic regulation, the flow of cerebrospinal fluid, and the subsequent dispersion of signalling molecules via the co-ordinated beating of their cilia. Deficits to ependymal cell development and function have been implicated in the formation of hydrocephalus, but the transcriptional mechanisms underpinning ependymal development remain poorly characterised. Findings Here, we demonstrate that the transcription factor nuclear factor IX (NFIX) plays a central role in the development of the ependymal cell layer of the lateral ventricles. Expression of ependymal cell-specific markers is delayed in the absence of Nfix. Moreover, Nfix-deficient mice exhibit aberrant ependymal cell morphology at postnatal day 15, culminating in abnormal thickening and intermittent loss of this cell layer. Finally, we reveal Foxj1, a key factor promoting ependymal cell maturation, as a target for NFIX-mediated transcriptional activation. Conclusions Collectively, our data indicate that ependymal cell development is reliant, at least in part, on NFIX expression, further implicating this transcription factor as a mediator of multiple aspects of radial glial biology during corticogenesis. Electronic supplementary material The online version of this article (10.1186/s13064-018-0099-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Diana Vidovic
- The School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Raul Ayala Davila
- The School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Richard M Gronostajski
- Department of Biochemistry, Program in Genetics, Genomics and Bioinformatics, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, New York, 14260, USA
| | - Tracey J Harvey
- The School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Michael Piper
- The School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia. .,Queensland Brain Institute, The University of Queensland, Brisbane, 4072, Australia.
| |
Collapse
|
28
|
Abstract
A hypercaloric diet combined with a sedentary lifestyle is a major risk factor for the development of insulin resistance, type 2 diabetes mellitus (T2DM) and associated comorbidities. Standard treatment for T2DM begins with lifestyle modification, and includes oral medications and insulin therapy to compensate for progressive β-cell failure. However, current pharmaceutical options for T2DM are limited in that they do not maintain stable, durable glucose control without the need for treatment intensification. Furthermore, each medication is associated with adverse effects, which range from hypoglycaemia to weight gain or bone loss. Unexpectedly, fibroblast growth factor 1 (FGF1) and its low mitogenic variants have emerged as potentially safe candidates for restoring euglycaemia, without causing overt adverse effects. In particular, a single peripheral injection of FGF1 can lower glucose to normal levels within hours, without the risk of hypoglycaemia. Similarly, a single intracerebroventricular injection of FGF1 can induce long-lasting remission of the diabetic phenotype. This Review discusses potential mechanisms by which centrally administered FGF1 improves central glucose-sensing and peripheral glucose uptake in a sustained manner. Specifically, we explore the potential crosstalk between FGF1 and glucose-sensing neuronal circuits, hypothalamic neural stem cells and synaptic plasticity. Finally, we highlight therapeutic considerations of FGF1 and compare its metabolic actions with FGF15 (rodents), FGF19 (humans) and FGF21.
Collapse
Affiliation(s)
- Emanuel Gasser
- Gene Expression Laboratory, Salk Institute for Biological Studies
| | - Christopher P Moutos
- Gene Expression Laboratory, Salk Institute for Biological Studies
- Howard Hughes Medical Institute, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California 92037, USA
- College of Medicine, University of Arkansas for Medical Sciences, 4301 West Markham Street, Little Rock, Arkansas 72205, USA
| | - Michael Downes
- Gene Expression Laboratory, Salk Institute for Biological Studies
| | - Ronald M Evans
- Gene Expression Laboratory, Salk Institute for Biological Studies
- Howard Hughes Medical Institute, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California 92037, USA
| |
Collapse
|
29
|
Narla ST, Lee YW, Benson C, Sarder P, Brennand K, Stachowiak E, Stachowiak M. Common developmental genome deprogramming in schizophrenia - Role of Integrative Nuclear FGFR1 Signaling (INFS). Schizophr Res 2017; 185:17-32. [PMID: 28094170 PMCID: PMC5507209 DOI: 10.1016/j.schres.2016.12.012] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 12/06/2016] [Accepted: 12/12/2016] [Indexed: 12/16/2022]
Abstract
The watershed-hypothesis of schizophrenia asserts that over 200 different mutations dysregulate distinct pathways that converge on an unspecified common mechanism(s) that controls disease ontogeny. Consistent with this hypothesis, our RNA-sequencing of neuron committed cells (NCCs) differentiated from established iPSCs of 4 schizophrenia patients and 4 control subjects uncovered a dysregulated transcriptome of 1349 mRNAs common to all patients. Data reveals a global dysregulation of developmental genome, deconstruction of coordinated mRNA networks, and the formation of aberrant, new coordinated mRNA networks indicating a concerted action of the responsible factor(s). Sequencing of miRNA transcriptomes demonstrated an overexpression of 16 miRNAs and deconstruction of interactive miRNA-mRNA networks in schizophrenia NCCs. ChiPseq revealed that the nuclear (n) form of FGFR1, a pan-ontogenic regulator, is overexpressed in schizophrenia NCCs and overtargets dysregulated mRNA and miRNA genes. The nFGFR1 targeted 54% of all human gene promoters and 84.4% of schizophrenia dysregulated genes. The upregulated genes reside within major developmental pathways that control neurogenesis and neuron formation, whereas downregulated genes are involved in oligodendrogenesis. Our results indicate (i) an early (preneuronal) genomic etiology of schizophrenia, (ii) dysregulated genes and new coordinated gene networks are common to unrelated cases of schizophrenia, (iii) gene dysregulations are accompanied by increased nFGFR1-genome interactions, and (iv) modeling of increased nFGFR1 by an overexpression of a nFGFR1 lead to up or downregulation of selected genes as observed in schizophrenia NCCs. Together our results designate nFGFR1 signaling as a potential common dysregulated mechanism in investigated patients and potential therapeutic target in schizophrenia.
Collapse
Affiliation(s)
- S. T. Narla
- Department of Pathology and Anatomical Sciences, State University of New York at Buffalo, Buffalo, NY, USA,Western New York Stem Cell Culture and Analysis Center, State University of New York at Buffalo, Buffalo, NY, USA
| | - Y-W. Lee
- Department of Pathology and Anatomical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - C.A. Benson
- Department of Pathology and Anatomical Sciences, State University of New York at Buffalo, Buffalo, NY, USA,Western New York Stem Cell Culture and Analysis Center, State University of New York at Buffalo, Buffalo, NY, USA
| | - P. Sarder
- Department of Pathology and Anatomical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - K. Brennand
- Icahn School of Medicine at Mount Sinai, Departments of Psychiatry and Neuroscience, New York, NY, USA
| | - E.K. Stachowiak
- Department of Pathology and Anatomical Sciences, State University of New York at Buffalo, Buffalo, NY, USA,Western New York Stem Cell Culture and Analysis Center, State University of New York at Buffalo, Buffalo, NY, USA
| | - M.K. Stachowiak
- Department of Pathology and Anatomical Sciences, State University of New York at Buffalo, Buffalo, NY, USA,Western New York Stem Cell Culture and Analysis Center, State University of New York at Buffalo, Buffalo, NY, USA,Correspondence should be addressed to Michal K. Stachowiak Department of Pathology and Anatomical Sciences, SUNY, 3435 Main Street, 206A Farber Hall, Buffalo, N.Y. 14214, tel. (716) 829 3540
| |
Collapse
|
30
|
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
|
31
|
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
|
32
|
The Function of FGFR1 Signalling in the Spinal Cord: Therapeutic Approaches Using FGFR1 Ligands after Spinal Cord Injury. Neural Plast 2017; 2017:2740768. [PMID: 28197342 PMCID: PMC5286530 DOI: 10.1155/2017/2740768] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 12/25/2016] [Indexed: 11/24/2022] Open
Abstract
Extensive research is ongoing that concentrates on finding therapies to enhance CNS regeneration after spinal cord injury (SCI) and to cure paralysis. This review sheds light on the role of the FGFR pathway in the injured spinal cord and discusses various therapies that use FGFR activating ligands to promote regeneration after SCI. We discuss studies that use peripheral nerve grafts or Schwann cell grafts in combination with FGF1 or FGF2 supplementation. Most of these studies show evidence that these therapies successfully enhance axon regeneration into the graft. Further they provide evidence for partial recovery of sensory function shown by electrophysiology and motor activity evidenced by behavioural data. We also present one study that indicates that combination with additional, synergistic factors might further drive the system towards functional regeneration. In essence, this review summarises the potential of nerve and cell grafts combined with FGF1/2 supplementation to improve outcome even after severe spinal cord injury.
Collapse
|
33
|
Spasova MS, Chen X, Sadowska GB, Horton ER, Lim YP, Stonestreet BS. Ischemia reduces inter-alpha inhibitor proteins in the brain of the ovine fetus. Dev Neurobiol 2016; 77:726-737. [PMID: 27618403 DOI: 10.1002/dneu.22451] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 09/02/2016] [Accepted: 09/08/2016] [Indexed: 01/04/2023]
Abstract
Hypoxic-ischemic (HI) brain injury is a major cause of neurological abnormalities in the perinatal period. Inflammation contributes to the evolution of HI brain injury. Inter-alpha inhibitor proteins (IAIPs) are a family of proteins that are part of the innate immune system. We have reported that endogenous IAIPs exhibit developmental changes in ovine brain and that exogenous IAIP treatment reduces neuronal death in HI neonatal rats. However, the effects of HI on endogenous IAIPs in brain have not been previously examined. In this study, we examined the effects of ischemia-reperfusion on endogenous IAIPs levels in fetal sheep brain. Cerebral cortex, cerebellum, cervical spinal cord, choroid plexus, and CSF were snap frozen from sham control fetuses at 127 days gestation and after 30-min of carotid occlusion and 4-, 24-, and 48-h of reperfusion. IAIP levels were determined by Western immunoblot. IAIP expressions of the 250 kDa Inter-alpha inhibitor (IaI) and 125 kDa Pre-alpha inhibitor (PaI) in cerebral cortex and PaI in cerebellum were reduced (p < 0.05) 4-h after ischemia compared with controls and returned toward control levels 24- and 48-h after ischemia. CSF PaI and IaI were reduced 48 h after ischemia. We conclude that IAIPs in cerebral cortex and cerebellum are reduced by brain ischemia, and return toward control levels between 24 and 48 h after ischemia. However, changes in CSF IAIPs were delayed, exhibiting decreases 48 h after ischemia. We speculate that the decreases in endogenous IAIPs reflect increased utilization, potentially suggesting that they have endogenous neuroprotective properties. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 77: 726-737, 2017.
Collapse
Affiliation(s)
- Mariya S Spasova
- Department of Pediatrics, the Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, RI, 02905
| | - Xiaodi Chen
- Department of Pediatrics, the Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, RI, 02905
| | - Grazyna B Sadowska
- Department of Pediatrics, the Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, RI, 02905
| | - Edward R Horton
- Department of Pediatrics, the Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, RI, 02905
| | - Yow-Pin Lim
- ProThera Biologics, Inc, Providence, RI, 02903
| | - Barbara S Stonestreet
- Department of Pediatrics, the Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, RI, 02905
| |
Collapse
|
34
|
Litvin Y, Turner CA, Rios MB, Maras PM, Chaudhury S, Baker MR, Blandino P, Watson SJ, Akil H, McEwen B. Fibroblast growth factor 2 alters the oxytocin receptor in a developmental model of anxiety-like behavior in male rat pups. Horm Behav 2016; 86:64-70. [PMID: 27693608 PMCID: PMC5789801 DOI: 10.1016/j.yhbeh.2016.09.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 09/21/2016] [Accepted: 09/26/2016] [Indexed: 12/21/2022]
Abstract
We aimed to determine the short-term effects of early-life stress in the form of maternal separation (MS) on anxiety-like behavior in male rat pups. In order to assess anxiety, we measured 40kHz separation-induced ultrasonic vocalizations (USV) on postnatal day (PND) 11. We further aimed to evaluate the potential involvement of two neurochemical systems known to regulate social and anxiety-like behaviors throughout life: oxytocin (OT) and fibroblast growth factor 2 (FGF2). For these purposes, we tested the effects of neonatal administration (on PND1) of an acute dose of FGF2 on USV and its potential interaction with MS. In addition, we validated the anxiolytic effects of OT and measured oxytocin receptor (OTR) gene expression, binding and epigenetic regulation via histone acetylation. Our results show that MS potentiated USV while acute administration of OT and FGF2 attenuated them. Further, we found that both FGF2 and MS increased OTR gene expression and the association of acH3K14 with the OTR promoter in the bed nucleus of the stria terminalis (BNST). Comparable changes, though not as pronounced, were also found for the central amygdala (CeA). Our findings suggest that FGF2 may exert its anxiolytic effects in male MS rats by a compensatory increase in the acetylation of the OTR promoter to overcome reduced OT levels in the BNST.
Collapse
Affiliation(s)
- Yoav Litvin
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, United States
| | - Cortney A Turner
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, United States
| | - Mariel B Rios
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, United States
| | - Pamela M Maras
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, United States
| | - Sraboni Chaudhury
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, United States
| | - Miriam R Baker
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, United States
| | - Peter Blandino
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, United States
| | - Stanley J Watson
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, United States
| | - Huda Akil
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, United States
| | - Bruce McEwen
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, United States.
| |
Collapse
|
35
|
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
|
36
|
Abstract
Hippocampal area CA2 has several features that distinguish it from CA1 and CA3, including a unique gene expression profile, failure to display long-term potentiation and relative resistance to cell death. A recent increase in interest in the CA2 region, combined with the development of new methods to define and manipulate its neurons, has led to some exciting new discoveries on the properties of CA2 neurons and their role in behaviour. Here, we review these findings and call attention to the idea that the definition of area CA2 ought to be revised in light of gene expression data.
Collapse
|
37
|
Kaur C, Rathnasamy G, Ling EA. The Choroid Plexus in Healthy and Diseased Brain. J Neuropathol Exp Neurol 2016; 75:198-213. [DOI: 10.1093/jnen/nlv030] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
|
38
|
Huang C, Yuan P, Wu J, Huang J. Estrogen regulates excitatory amino acid carrier 1 (EAAC1) expression through sphingosine kinase 1 (SphK1) transacting FGFR-mediated ERK signaling in rat C6 astroglial cells. Neuroscience 2016; 319:9-22. [PMID: 26804240 DOI: 10.1016/j.neuroscience.2016.01.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 01/04/2016] [Accepted: 01/12/2016] [Indexed: 12/28/2022]
Abstract
Excitatory amino acid carrier 1 (EAAC1) is one important subtype of the excitatory amino acid transporters (EAATs), and its absence can increase the vulnerability to oxidative stress in neural tissue. Enhanced expression of EAAC1 can provide neuroprotection in multiple disorders, including ischemia and multiple sclerosis. However, the mechanism regulating EAAC1 expression is not fully understood. Using rat C6 astroglial cells, which specifically express EAAC1, we found that 17β-estradiol (E2) and (±)-1-[(3aR(∗),4S(∗),9bS(∗))-4-(6-bromo-1,3-benzodioxol-5-yl)-3a,4,5,9b-tetrahydro-3H-cyclopenta[c]quinolin-8-yl]-ethanone (G1), an agonist of the G-protein-coupled estrogen receptor (GPR30), strongly increased EAAC1 protein levels and protected cells from hydrogen peroxide (H2O2) toxicity. We further found that E2/G1 activated sphingosine kinase 1 (SphK1) via GPR30, resulting in the transcription of fibroblast growth factor 2 (FGF2), which stimulated its receptor (FGFR) and led to the phosphorylation of FGFR substrate 2α (FRS2α). This triggered downstream ERK1/2 signaling for the expression of EAAC1. Both the knockdown of FGF2 by siRNA and the pharmacological suppression of the FGFR-ERK cascade abolished the E2/G1 effect on EAAC1 expression. Overall, our work characterizes a signaling pathway by which E2 transactivates FGFR-ERK to induce EAAC1 expression in an FGF2-dependent manner. This occurs through SphK1 activation via GPR30 and leads to a resistance to H2O2 toxicity. This signal transduction pathway may provide novel insights into our understanding of the neuroprotective effects of E2 and may reveal new therapeutic targets or drugs for regulating the oxidative toxicity effects of various neurological diseases.
Collapse
Affiliation(s)
- C Huang
- College of Life Science, Wuhan University, Wuhan 430072, PR China
| | - P Yuan
- Department of Neurobiology, Yale University School of Medicine, New Haven, CT, USA
| | - J Wu
- College of Life Science, Wuhan University, Wuhan 430072, PR China
| | - J Huang
- College of Life Science, Wuhan University, Wuhan 430072, PR China.
| |
Collapse
|
39
|
Elsayed M, Magistretti PJ. A New Outlook on Mental Illnesses: Glial Involvement Beyond the Glue. Front Cell Neurosci 2015; 9:468. [PMID: 26733803 PMCID: PMC4679853 DOI: 10.3389/fncel.2015.00468] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 11/16/2015] [Indexed: 12/27/2022] Open
Abstract
Mental illnesses have long been perceived as the exclusive consequence of abnormalities in neuronal functioning. Until recently, the role of glial cells in the pathophysiology of mental diseases has largely been overlooked. However recently, multiple lines of evidence suggest more diverse and significant functions of glia with behavior-altering effects. The newly ascribed roles of astrocytes, oligodendrocytes and microglia have led to their examination in brain pathology and mental illnesses. Indeed, abnormalities in glial function, structure and density have been observed in postmortem brain studies of subjects diagnosed with mental illnesses. In this review, we discuss the newly identified functions of glia and highlight the findings of glial abnormalities in psychiatric disorders. We discuss these preclinical and clinical findings implicating the involvement of glial cells in mental illnesses with the perspective that these cells may represent a new target for treatment.
Collapse
Affiliation(s)
- Maha Elsayed
- Laboratory of Neuroenergetics and Cellular Dynamics, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne Lausanne, Switzerland
| | - Pierre J Magistretti
- Laboratory of Neuroenergetics and Cellular Dynamics, Brain Mind Institute, Ecole Polytechnique Fédérale de LausanneLausanne, Switzerland; Division of Biological and Environmental Sciences and Engineering, King Abdullah University of Science and TechnologyThuwal, Saudi Arabia; Department of Psychiatry, Center for Psychiatric Neurosciences, University of LausanneLausanne, Switzerland
| |
Collapse
|
40
|
Blocking Infralimbic Basic Fibroblast Growth Factor (bFGF or FGF2) Facilitates Extinction of Drug Seeking After Cocaine Self-Administration. Neuropsychopharmacology 2015; 40:2907-15. [PMID: 25994078 PMCID: PMC4864626 DOI: 10.1038/npp.2015.144] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Revised: 05/06/2015] [Accepted: 05/15/2015] [Indexed: 01/09/2023]
Abstract
Drug exposure results in structural and functional changes in brain regions that regulate reward and these changes may underlie the persistence of compulsive drug seeking and relapse. Neurotrophic factors, such as basic fibroblast growth factor (bFGF or FGF2), are necessary for neuronal survival, growth, and differentiation, and may contribute to these drug-induced changes. Following cocaine exposure, bFGF is increased in addiction-related brain regions, including the infralimbic medial prefrontal cortex (IL-mPFC). The IL-mPFC is necessary for extinction, but whether drug-induced overexpression of bFGF in this region affects extinction of drug seeking is unknown. Thus, we determined whether blocking bFGF in IL-mPFC would facilitate extinction following cocaine self-administration. Rats were trained to lever press for intravenous infusions of cocaine before extinction. Blocking bFGF in IL-mPFC before four extinction sessions resulted in facilitated extinction. In contrast, blocking bFGF alone was not sufficient to facilitate extinction, as blocking bFGF and returning rats to their home cage had no effect on subsequent extinction. Furthermore, bFGF protein expression increased in IL-mPFC following cocaine self-administration, an effect reversed by extinction. These results suggest that cocaine-induced overexpression of bFGF inhibits extinction, as blocking bFGF during extinction permits rapid extinction. Therefore, targeted reductions in bFGF during therapeutic interventions could enhance treatment outcomes for addiction.
Collapse
|
41
|
Kajitani N, Hisaoka-Nakashima K, Okada-Tsuchioka M, Hosoi M, Yokoe T, Morioka N, Nakata Y, Takebayashi M. Fibroblast growth factor 2 mRNA expression evoked by amitriptyline involves extracellular signal-regulated kinase-dependent early growth response 1 production in rat primary cultured astrocytes. J Neurochem 2015; 135:27-37. [DOI: 10.1111/jnc.13247] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 07/08/2015] [Accepted: 07/09/2015] [Indexed: 01/17/2023]
Affiliation(s)
- Naoto Kajitani
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences; Hiroshima University; Minami-ku Hiroshima Japan
- Division of Psychiatry and Neuroscience; Institute for Clinical Research; National Hospital Organization (NHO) Kure Medical Center and Chugoku Cancer Center; Kure Japan
| | - Kazue Hisaoka-Nakashima
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences; Hiroshima University; Minami-ku Hiroshima Japan
| | - Mami Okada-Tsuchioka
- Division of Psychiatry and Neuroscience; Institute for Clinical Research; National Hospital Organization (NHO) Kure Medical Center and Chugoku Cancer Center; Kure Japan
| | - Mayu Hosoi
- Division of Psychiatry and Neuroscience; Institute for Clinical Research; National Hospital Organization (NHO) Kure Medical Center and Chugoku Cancer Center; Kure Japan
| | - Toshiki Yokoe
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences; Hiroshima University; Minami-ku Hiroshima Japan
| | - Norimitsu Morioka
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences; Hiroshima University; Minami-ku Hiroshima Japan
| | - Yoshihiro Nakata
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences; Hiroshima University; Minami-ku Hiroshima Japan
| | - Minoru Takebayashi
- Division of Psychiatry and Neuroscience; Institute for Clinical Research; National Hospital Organization (NHO) Kure Medical Center and Chugoku Cancer Center; Kure Japan
- Department of Psychiatry; National Hospital Organization (NHO) Kure Medical Center and Chugoku Cancer Center; Kure Japan
| |
Collapse
|
42
|
Spasova MS, Sadowska GB, Threlkeld SW, Lim YP, Stonestreet BS. Ontogeny of inter-alpha inhibitor proteins in ovine brain and somatic tissues. Exp Biol Med (Maywood) 2015; 239:724-36. [PMID: 24728724 DOI: 10.1177/1535370213519195] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Inter-alpha inhibitor proteins (IAIPs) found in relatively high concentrations in human plasma are important in inflammation. IAIPs attenuate brain damage in young and adult subjects, decrease during sepsis and necrotizing enterocolitis in premature infants, and attenuate sepsis-related inflammation in newborn rats. Although a few studies have reported adult organ-specific IAIP expression, information is not available on age-dependent IAIP expression. Given evidence suggesting IAIPs attenuate brain damage in young and adult subjects, and inflammation in newborns, we examined IAIP expression in plasma, cerebral cortex (CC), choroid plexus (CP), cerebral spinal fluid (CSF), and somatic organs in fetal, newborn, and adult sheep to determine the endogenous expression patterns of these proteins during development. IAIPs (enzyme-linked immunosorbent assay) were higher in newborn and adult than fetal plasma (P < 0.05). Western immunoblot detected 125 kDa PaI (Pre-alpha Inhibitor) and 250 kDa IaI (Inter-alpha Inhibitor) in plasma, CNS, and somatic organs. PaI expression in CC and CP was higher in fetuses than newborns and adults, but IaI expression was higher in adults than fetuses and newborns. Both PaI and IaI were higher in fetal than newborn CSF. IAIPs exhibited organ-specific ontogenic patterns in placenta, liver, heart, and kidney. These results provide evidence for the first time that plasma, brain, placenta, liver, heart, and kidney express IAIPs throughout ovine development and that expression patterns are unique to each organ. Although exact functions of IAIPs in CNS and somatic tissues are not known, their presence in relatively high amounts during development suggests their potential importance in brain and organ development.
Collapse
|
43
|
Terranova C, Narla ST, Lee YW, Bard J, Parikh A, Stachowiak EK, Tzanakakis ES, Buck MJ, Birkaya B, Stachowiak MK. Global Developmental Gene Programing Involves a Nuclear Form of Fibroblast Growth Factor Receptor-1 (FGFR1). PLoS One 2015; 10:e0123380. [PMID: 25923916 PMCID: PMC4414453 DOI: 10.1371/journal.pone.0123380] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 02/17/2015] [Indexed: 12/11/2022] Open
Abstract
Genetic studies have placed the Fgfr1 gene at the top of major ontogenic pathways that enable gastrulation, tissue development and organogenesis. Using genome-wide sequencing and loss and gain of function experiments the present investigation reveals a mechanism that underlies global and direct gene regulation by the nuclear form of FGFR1, ensuring that pluripotent Embryonic Stem Cells differentiate into Neuronal Cells in response to Retinoic Acid. Nuclear FGFR1, both alone and with its partner nuclear receptors RXR and Nur77, targets thousands of active genes and controls the expression of pluripotency, homeobox, neuronal and mesodermal genes. Nuclear FGFR1 targets genes in developmental pathways represented by Wnt/β-catenin, CREB, BMP, the cell cycle and cancer-related TP53 pathway, neuroectodermal and mesodermal programing networks, axonal growth and synaptic plasticity pathways. Nuclear FGFR1 targets the consensus sequences of transcription factors known to engage CREB-binding protein, a common coregulator of transcription and established binding partner of nuclear FGFR1. This investigation reveals the role of nuclear FGFR1 as a global genomic programmer of cell, neural and muscle development.
Collapse
Affiliation(s)
- Christopher Terranova
- Department of Pathology and Anatomical Sciences, Western New York Stem Cell Culture and Analysis Center, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Sridhar T. Narla
- Department of Pathology and Anatomical Sciences, Western New York Stem Cell Culture and Analysis Center, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Yu-Wei Lee
- Department of Pathology and Anatomical Sciences, Western New York Stem Cell Culture and Analysis Center, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Jonathan Bard
- Next-Generation Sequencing and Expression Analysis Core, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Abhirath Parikh
- Department of Chemical and Biological Engineering, Western New York Stem Cell Culture and Analysis Center, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Ewa K. Stachowiak
- Department of Pathology and Anatomical Sciences, Western New York Stem Cell Culture and Analysis Center, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Emmanuel S. Tzanakakis
- Department of Chemical and Biological Engineering, Western New York Stem Cell Culture and Analysis Center, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Michael J. Buck
- Department of Biochemistry, Genomics and Bioinformatics Core, Western New York Stem Cell Culture and Analysis Center, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Barbara Birkaya
- Department of Pathology and Anatomical Sciences, Western New York Stem Cell Culture and Analysis Center, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Michal K. Stachowiak
- Department of Pathology and Anatomical Sciences, Western New York Stem Cell Culture and Analysis Center, State University of New York at Buffalo, Buffalo, New York, United States of America
- * E-mail:
| |
Collapse
|
44
|
Stachowiak MK, Birkaya B, Aletta JM, Narla ST, Benson CA, Decker B, Stachowiak EK. "Nuclear FGF receptor-1 and CREB binding protein: an integrative signaling module". J Cell Physiol 2015; 230:989-1002. [PMID: 25503065 DOI: 10.1002/jcp.24879] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 12/05/2014] [Indexed: 12/15/2022]
Abstract
In this review we summarize the current understanding of a novel integrative function of Fibroblast Growth Factor Receptor-1 (FGFR1) and its partner CREB Binding Protein (CBP) acting as a nuclear regulatory complex. Nuclear FGFR1 and CBP interact with and regulate numerous genes on various chromosomes. FGFR1 dynamic oscillatory interactions with chromatin and with specific genes, underwrites gene regulation mediated by diverse developmental signals. Integrative Nuclear FGFR1 Signaling (INFS) effects the differentiation of stem cells and neural progenitor cells via the gene-controlling Feed-Forward-And-Gate mechanism. Nuclear accumulation of FGFR1 occurs in numerous cell types and disruption of INFS may play an important role in developmental disorders such as schizophrenia, and in metastatic diseases such as cancer. Enhancement of INFS may be used to coordinate the gene regulation needed to activate cell differentiation for regenerative purposes or to provide interruption of cancer stem cell proliferation.
Collapse
Affiliation(s)
- Michal K Stachowiak
- Department of Pathology and Anatomical Sciences, Western New York Stem Cells Culture and Analysis Center, State University of New York, Buffalo
| | | | | | | | | | | | | |
Collapse
|
45
|
Liang Q, Zhong L, Zhang J, Wang Y, Bornstein SR, Triggle CR, Ding H, Lam KSL, Xu A. FGF21 maintains glucose homeostasis by mediating the cross talk between liver and brain during prolonged fasting. Diabetes 2014; 63:4064-75. [PMID: 25024372 DOI: 10.2337/db14-0541] [Citation(s) in RCA: 208] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Hepatic gluconeogenesis is a main source of blood glucose during prolonged fasting and is orchestrated by endocrine and neural pathways. Here we show that the hepatocyte-secreted hormone fibroblast growth factor 21 (FGF21) induces fasting gluconeogenesis via the brain-liver axis. Prolonged fasting induces activation of the transcription factor peroxisome proliferator-activated receptor α (PPARα) in the liver and subsequent hepatic production of FGF21, which enters into the brain to activate the hypothalamic-pituitary-adrenal (HPA) axis for release of corticosterone, thereby stimulating hepatic gluconeogenesis. Fasted FGF21 knockout (KO) mice exhibit severe hypoglycemia and defective hepatic gluconeogenesis due to impaired activation of the HPA axis and blunted release of corticosterone, a phenotype similar to that observed in PPARα KO mice. By contrast, intracerebroventricular injection of FGF21 reverses fasting hypoglycemia and impairment in hepatic gluconeogenesis by restoring corticosterone production in both FGF21 KO and PPARα KO mice, whereas all these central effects of FGF21 were abrogated by blockage of hypothalamic FGF receptor-1. FGF21 acts directly on the hypothalamic neurons to activate the mitogen-activated protein kinase extracellular signal-related kinase 1/2 (ERK1/2), thereby stimulating the expression of corticotropin-releasing hormone by activation of the transcription factor cAMP response element binding protein. Therefore, FGF21 maintains glucose homeostasis during prolonged fasting by fine tuning the interorgan cross talk between liver and brain.
Collapse
Affiliation(s)
- Qingning Liang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ling Zhong
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jialiang Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yu Wang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China
| | | | - Chris R Triggle
- Department of Pharmacology, Weill Cornell Medical College in Qatar, Doha, Qatar
| | - Hong Ding
- Department of Pharmacology, Weill Cornell Medical College in Qatar, Doha, Qatar
| | - Karen S L Lam
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China Department of Medicine, The University of Hong Kong, Hong Kong, China Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
46
|
Lelliott CJ, Ahnmark A, Admyre T, Ahlstedt I, Irving L, Keyes F, Patterson L, Mumphrey MB, Bjursell M, Gorman T, Bohlooly-Y M, Buchanan A, Harrison P, Vaughan T, Berthoud HR, Lindén D. Monoclonal antibody targeting of fibroblast growth factor receptor 1c ameliorates obesity and glucose intolerance via central mechanisms. PLoS One 2014; 9:e112109. [PMID: 25427253 PMCID: PMC4245083 DOI: 10.1371/journal.pone.0112109] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 10/13/2014] [Indexed: 12/21/2022] Open
Abstract
We have generated a novel monoclonal antibody targeting human FGFR1c (R1c mAb) that caused profound body weight and body fat loss in diet-induced obese mice due to decreased food intake (with energy expenditure unaltered), in turn improving glucose control. R1c mAb also caused weight loss in leptin-deficient ob/ob mice, leptin receptor-mutant db/db mice, and in mice lacking either the melanocortin 4 receptor or the melanin-concentrating hormone receptor 1. In addition, R1c mAb did not change hypothalamic mRNA expression levels of Agrp, Cart, Pomc, Npy, Crh, Mch, or Orexin, suggesting that R1c mAb could cause food intake inhibition and body weight loss via other mechanisms in the brain. Interestingly, peripherally administered R1c mAb accumulated in the median eminence, adjacent arcuate nucleus and in the circumventricular organs where it activated the early response gene c-Fos. As a plausible mechanism and coinciding with the initiation of food intake suppression, R1c mAb induced hypothalamic expression levels of the cytokines Monocyte chemoattractant protein 1 and 3 and ERK1/2 and p70 S6 kinase 1 activation.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/pharmacology
- Arcuate Nucleus of Hypothalamus/drug effects
- Arcuate Nucleus of Hypothalamus/metabolism
- Arcuate Nucleus of Hypothalamus/physiopathology
- Chemokine CCL2/agonists
- Chemokine CCL2/genetics
- Chemokine CCL2/metabolism
- Chemokine CCL7/agonists
- Chemokine CCL7/genetics
- Chemokine CCL7/metabolism
- Circumventricular Organs/drug effects
- Circumventricular Organs/metabolism
- Circumventricular Organs/physiopathology
- Eating/drug effects
- Energy Metabolism
- Female
- Gene Expression Regulation
- Glucose Intolerance/drug therapy
- Glucose Intolerance/genetics
- Glucose Intolerance/metabolism
- Glucose Intolerance/physiopathology
- Humans
- Hypothalamus/drug effects
- Hypothalamus/metabolism
- Hypothalamus/physiopathology
- Leptin/deficiency
- Leptin/genetics
- Mice
- Mice, Knockout
- Mice, Obese
- Mitogen-Activated Protein Kinases/genetics
- Mitogen-Activated Protein Kinases/metabolism
- Obesity/drug therapy
- Obesity/genetics
- Obesity/metabolism
- Obesity/physiopathology
- Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Receptor, Fibroblast Growth Factor, Type 1/metabolism
- Receptor, Melanocortin, Type 4/deficiency
- Receptor, Melanocortin, Type 4/genetics
- Receptors, Somatostatin/deficiency
- Receptors, Somatostatin/genetics
- Ribosomal Protein S6 Kinases, 70-kDa/genetics
- Ribosomal Protein S6 Kinases, 70-kDa/metabolism
- Serum Response Factor/agonists
- Serum Response Factor/genetics
- Serum Response Factor/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Christopher J. Lelliott
- Cardiovascular & Metabolic Disease Innovative Medicines, Dept of Bioscience Diabetes, AstraZeneca, Mölndal, Sweden
| | - Andrea Ahnmark
- Cardiovascular & Metabolic Disease Innovative Medicines, Dept of Bioscience Diabetes, AstraZeneca, Mölndal, Sweden
| | - Therese Admyre
- Discovery Sciences Transgenics, AstraZeneca, Mölndal, Sweden
| | - Ingela Ahlstedt
- Cardiovascular & Metabolic Disease Innovative Medicines, Dept of Bioscience Diabetes, AstraZeneca, Mölndal, Sweden
| | - Lorraine Irving
- Antibody Discovery and Protein Engineering, MedImmune, Cambridge, United Kingdom
| | - Feenagh Keyes
- Antibody Discovery and Protein Engineering, MedImmune, Cambridge, United Kingdom
| | - Laurel Patterson
- Neurobiology of Nutrition Laboratory, Pennington Biomedical Research Center, Baton Rouge, United States of America
| | - Michael B. Mumphrey
- Neurobiology of Nutrition Laboratory, Pennington Biomedical Research Center, Baton Rouge, United States of America
| | - Mikael Bjursell
- Discovery Sciences Transgenics, AstraZeneca, Mölndal, Sweden
| | - Tracy Gorman
- AstraZeneca, Discovery Sciences, Mereside, Alderley Park, Macclesfield, Cheshire, United Kingdom
| | | | - Andrew Buchanan
- Antibody Discovery and Protein Engineering, MedImmune, Cambridge, United Kingdom
| | - Paula Harrison
- Antibody Discovery and Protein Engineering, MedImmune, Cambridge, United Kingdom
| | - Tristan Vaughan
- Antibody Discovery and Protein Engineering, MedImmune, Cambridge, United Kingdom
| | - Hans-Rudolf Berthoud
- Neurobiology of Nutrition Laboratory, Pennington Biomedical Research Center, Baton Rouge, United States of America
| | - Daniel Lindén
- Cardiovascular & Metabolic Disease Innovative Medicines, Dept of Bioscience Diabetes, AstraZeneca, Mölndal, Sweden
- * E-mail:
| |
Collapse
|
47
|
Smith KM, Maragnoli ME, Phull PM, Tran KM, Choubey L, Vaccarino FM. Fgfr1 inactivation in the mouse telencephalon results in impaired maturation of interneurons expressing parvalbumin. PLoS One 2014; 9:e103696. [PMID: 25116473 PMCID: PMC4130531 DOI: 10.1371/journal.pone.0103696] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 07/04/2014] [Indexed: 11/18/2022] Open
Abstract
Fibroblast growth factors (Fgfs) and their receptors (Fgfr) are expressed in the developing and adult CNS. Previous studies demonstrated a decrease in cortical interneurons and locomotor hyperactivity in mice with a conditional Fgfr1 deletion generated in radial glial cells during midneurogenesis (Fgfr1f/f;hGfapCre+). Here, we report earlier and more extensive inactivation of Fgfr1 in neuroepithelial cells of the CNS (Fgfr1f/f;NesCre+). Similar to findings in Fgfr1f/f;hGfapCre+ mice, parvalbumin positive (PV+) cortical interneurons are also decreased in the neocortex of Fgfr1f/f;NesCre+ mice when compared to control littermates (Fgfr1f/f). Fgfr1f/f;NesCre+ embryos do not differ from controls in the initial specification of GABAergic cells in the ganglionic eminence (GE) as assessed by in situ hybridization for Dlx2, Mash1 and Nkx2. Equal numbers of GABAergic neuron precursors genetically labeled with green fluorescent protein (GFP) were observed at P0 in Fgfr1f/f;hGfapCre+;Gad1-GFP mutant mice. However, fewer GFP+ and GFP+/PV+ interneurons were observed in these mutants at adulthood, indicating that a decrease in cortical interneuron markers is occurring postnatally. Fgfr1 is expressed in cortical astrocytes in the postnatal brain. To test whether the astrocytes of mice lacking Fgfr1 are less capable of supporting interneurons, we co-cultured wild type Gad1-GFP+ interneuron precursors isolated from the medial GE (MGE) with astrocytes from Fgfr1f/f control or Fgfr1f/f;hGfapCre+ mice. Interneurons grown on Fgfr1 deficient astrocytes had small soma size and fewer neurites per cell, but no differences in cell survival. Decreased soma size of Gad67 immunopositive interneurons was also observed in the cortex of adult Fgfr1f/f;NesCre+ mice. Our data indicate that astrocytes from Fgfr1 mutants are impaired in supporting the maturation of cortical GABAergic neurons in the postnatal period. This model may elucidate potential mechanisms of impaired PV interneuron maturation relevant to neuropsychiatric disorders that develop in childhood and adolescence.
Collapse
Affiliation(s)
- Karen Müller Smith
- Child Study Center, Yale University, New Haven, Connecticut, United States of America
| | | | - Pooja M. Phull
- Child Study Center, Yale University, New Haven, Connecticut, United States of America
| | - Kathy May Tran
- Child Study Center, Yale University, New Haven, Connecticut, United States of America
| | - Lisha Choubey
- Department of Biology, University of Louisiana at Lafayette, Lafayette, Louisiana, United States of America
| | - Flora M. Vaccarino
- Child Study Center, Yale University, New Haven, Connecticut, United States of America
- Department of Neurobiology, Yale University, New Haven, Connecticut, United States of America
- Kavli Institute for Neuroscience, Yale University, New Haven, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
48
|
He S, Zhang T, Hong B, Peng D, Su H, Lin Z, Fang Y, Jiang K, Liu X, Li H. Decreased serum fibroblast growth factor - 2 levels in pre- and post-treatment patients with major depressive disorder. Neurosci Lett 2014; 579:168-72. [PMID: 25079902 DOI: 10.1016/j.neulet.2014.07.035] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Revised: 07/18/2014] [Accepted: 07/20/2014] [Indexed: 12/24/2022]
Abstract
Increasing evidence indicates that neurotrophic factor dysfunction might be involved in the pathophysiology and treatment of major depressive disorder (MDD). Fibroblast growth factor (FGF)-2, one of the major neurotrophins, plays an important role in the central nervous system (CNS). The aim of this study was to explore whether the FGF-2 in serum was associated with MDD and to evaluate the effects of antidepressant treatment on serum FGF-2 levels. Serum FGF-2 levels were determined in 28 pre- and post-treatment MDD patients and 30 healthy controls using ELISA. The results of the current study revealed that serum FGF-2 levels in MDD patients were significantly lower than those in healthy controls (p=0.005), and the serum FGF-2 levels decreased significantly but marginally following treatment for 8 weeks (p=0.005). These findings demonstrate that the lower serum FGF-2 levels contribute to the pathophysiology of MDD and that FGF-2 may be used as a peripheral biological marker for MDD.
Collapse
Affiliation(s)
- Shen He
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Tianhong Zhang
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Bo Hong
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Daihui Peng
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Hui Su
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Zhiguang Lin
- Biochemistry Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Yiru Fang
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Kaida Jiang
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Xiaohua Liu
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| | - Huafang Li
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; Biochemistry Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; Medical Institution Conducting Clinical Trials for Human Used Drug, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| |
Collapse
|
49
|
PEGylated rhFGF-2 Conveys Long-term Neuroprotection and Improves Neuronal Function in a Rat Model of Parkinson’s Disease. Mol Neurobiol 2014; 51:32-42. [DOI: 10.1007/s12035-014-8750-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 05/08/2014] [Indexed: 10/25/2022]
|
50
|
Kang K, Lee SW, Han JE, Choi JW, Song MR. The complex morphology of reactive astrocytes controlled by fibroblast growth factor signaling. Glia 2014; 62:1328-44. [PMID: 24796693 DOI: 10.1002/glia.22684] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Revised: 03/19/2014] [Accepted: 04/17/2014] [Indexed: 12/12/2022]
Abstract
Astrocytes are the most abundant cell-type of the human brain and play a variety of roles in brain homeostasis and synaptic maturation, under normal conditions. However, astrocytes undergo dramatic pathological changes in response to brain injury, such as reactive gliosis and glial scar formation. Although abnormal hypertrophy and massive proliferation of astrocytes are obvious, the molecular identity and cues that dictate the structural changes in reactive astrocytes remain unclear. This study proposes that fibroblast growth factor (FGF) signaling is responsible for making astrocyte morphology more complex and hypertrophic in response to an inflammatory stimulus such as lipopolysaccharide. Primary astrocytes isolated from perinatal brains developed more branches in the presence of FGF8 or lesser branches in the presence of FGF2. Introduction of the constitutively active form of the FGF receptor 3 (caFGFR3) into the brain increases the structural complexity, with greater glial fibrillary acidic protein level in astrocytes, while overexpression of a dominant-negative form of FGFR3 (dnFGFR3) reduces it. Treatment of FGF8 facilitated the wound-healing process of primary astrocytes in vitro by changing their morphology, indicating that the FGF signal may control the responsiveness of astrocytes in injury conditions. Finally, the blockade of FGF signaling by introducing dnFGFR3 at the site of reactive gliosis reduces astrocyte branch formation and minimizes hypertrophic responses during reactive gliosis. Taken together, these results indicate that FGF8-FGFR3 signaling controls structural changes in astrocytes during reactive gliosis, under pathogenic conditions.
Collapse
Affiliation(s)
- Kyungjoon Kang
- School of Life Sciences, Bioimaging Research Center and Cell Dynamics Research Center, Gwangju Institute of Science and Technology, Oryong-dong, Buk-gu, Gwangju, 500-712, Republic of Korea
| | | | | | | | | |
Collapse
|