1
|
Cai X, Xie Z, Zhao J, Lu W, Zhu Z, Chen M, Huang Z, Ying Y, Fu Y, Xu J, Zhu S. FGF20 promotes spinal cord injury repair by inhibiting the formation of necrotic corpuscle P-MLKL/P-RIP1/P-RIP3 in neurons. J Cell Mol Med 2024; 28:e70109. [PMID: 39676730 DOI: 10.1111/jcmm.70109] [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: 11/23/2023] [Revised: 09/11/2024] [Accepted: 09/13/2024] [Indexed: 12/17/2024] Open
Abstract
The disruption of the local microenvironment subsequent to spinal cord injury (SCI) leads to a substantial loss of neurons in the affected region, which is a major contributing factor to impaired motor function recovery in patients. Fibroblast growth factor 20 (FGF20) is a neurotrophic factor that plays a crucial role in neuronal development and homeostasis. In this study, the recombinant human FGF20 (rhFGF20) was found to mitigate the process of necroptosis in a mouse model of SCI, thereby reducing neural functional deficits and promoting SCI repair. FGF20 protein was injected into the SCI mice via intraperitoneal injection. Using the BMS scale and inclined plane test, we found that FGF20 significantly promoted the recovery of motor function. The Nissl staining revealed the level of neuronal survival within the region of injury. The expression changes of NeuN, GAP43, NF200 and GFAP indicated that FGF20 has the nerve repair ability to delay the formation of glial scar. Through fluorescence detection of Ace-Tubulin and Tyr-Tubulin, FGF20 was revealed to promote the polymerization of axon-regenerated microtubules. Furthermore, FGF20 was also found to reduce the expression levels of necroptosis induced by SCI. These data suggest that FGF20 may exert a neuroprotective effect by inhibiting injury-induced necroptosis, thereby facilitating functional recovery following SCI. Moreover, systemic administration of FGF20 holds promise as a potential therapeutic strategy for repairing the damaged spinal cord. The discovery paves the way for a novel avenue of growth factor research in the field of SCI.
Collapse
Affiliation(s)
- Xiong Cai
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhenwen Xie
- The First Clinical School of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Juan Zhao
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wenjie Lu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhongwei Zhu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Min Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhiyang Huang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yibo Ying
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yining Fu
- The First Clinical School of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jiake Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Sipin Zhu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
2
|
Rahman ML, Bonnard AA, Wang F, Ruaud L, Guimiot F, Li Y, Defer I, Wang Y, Marchand V, Motorin Y, Yao B, Drunat S, Ghalei H. New ZNHIT3 Variants Disrupting snoRNP Assembly Cause Prenatal PEHO Syndrome with Isolated Hydrops. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.26.24312490. [PMID: 39252897 PMCID: PMC11383450 DOI: 10.1101/2024.08.26.24312490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
ZNHIT3 (zinc finger HIT type containing protein 3) is an evolutionarily conserved protein required for ribosome biogenesis by mediating the assembly of small nucleolar RNAs (snoRNAs) of class C/D into ribonucleoprotein complexes (snoRNPs). Missense mutations in the gene encoding ZNHIT3 protein have been previously reported to cause PEHO syndrome, a severe neurodevelopmental disorder typically presenting after birth. We discuss here the case of two fetuses from a single family who presented with isolated hydrops during the early second trimester of pregnancy, resulting in intrauterine demise. Autopsy revealed no associated malformation. Through whole-genome quartet analysis, we identified two novel variants within the ZNHIT3 gene, both inherited from healthy parents and occurring as compound heterozygotes in both fetuses. The c.40T>C p.Cys14Arg variant originated from the father, while the c.251_254delAAGA variant was of maternal origin. Analysis of the variants in human cell culture models reveals that both variants reduce cell growth, albeit to different extents, and impact the protein's stability and function in distinct ways. The c.251_254delAAGA results in production of a stable form of ZNHIT3 that lacks a domain required for mediating snoRNP biogenesis, whereas the c.40T>C p.Cys14Arg variation behaves similarly to the previously described PEHO-associated ZNHIT3 variants that destabilize the protein. Interestingly, both variations lead to a marked decrease in specific box C/D snoRNA levels, reduced rRNA levels and cellular translation. Analysis of rRNA methylation pattern in fetus samples reveals distinct sites of hypo 2'-O-methylation. RNA-seq analysis of undifferentiated and differentiated SHSY5Y cells transfected with the ZNHIT3 variants reveals differential expression of a set of genes, many of which are associated with developmental processes and RNA binding compared to cells expressing wild-type ZNHIT3. In summary, this work extends the phenotype of PEHO syndrome to include antenatal manifestations and describe the molecular defects induced by two novel ZNHIT3 variants.
Collapse
Affiliation(s)
- Md Lutfur Rahman
- Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Adeline A. Bonnard
- Département de Génétique, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France
- INSERM UMR 1131, Saint-Louis Research Institute, Paris University, Paris, France
| | - Feng Wang
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Lyse Ruaud
- Département de Génétique, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France
- INSERM UMR 1141, Paris-Cité University, NeuroDiderot, Paris, France
| | - Fabien Guimiot
- Département de Génétique, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France
- INSERM UMR 1141, Paris-Cité University, NeuroDiderot, Paris, France
| | - Yangping Li
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Ines Defer
- Département de Génétique, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France
- INSERM UMR 1131, Saint-Louis Research Institute, Paris University, Paris, France
| | - Yilin Wang
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Virginie Marchand
- Université de Lorraine, SMP IBSLor, Biopôle, 9 Avenue de la Forêt de Haye, Vandoeuvre-les-Nancy, France
| | - Yuri Motorin
- Université de Lorraine, UMR7365 IMoPA, CNRS, Biopôle, 9 Avenue de la Forêt de Haye, Vandoeuvre-les-Nancy, France
| | - Bing Yao
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Séverine Drunat
- Département de Génétique, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France
- INSERM UMR 1141, Paris-Cité University, NeuroDiderot, Paris, France
| | - Homa Ghalei
- Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
3
|
Dlugosova S, Spoutil F, Madureira Trufen CE, Melike Ogan B, Prochazkova M, Fedosieieva O, Nickl P, Aranaz Novaliches G, Sedlacek R, Prochazka J. Skeletal dysmorphology and mineralization defects in Fgf20 KO mice. Front Endocrinol (Lausanne) 2024; 15:1286365. [PMID: 39129916 PMCID: PMC11310068 DOI: 10.3389/fendo.2024.1286365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 07/02/2024] [Indexed: 08/13/2024] Open
Abstract
Introduction Fibroblast growth factor 20 (Fgf20), a member of the Fgf9 subfamily, was identified as an important regulator of bone differentiation and homeostasis processes. However, the role of Fgf20 in bone physiology has not been approached yet. Here we present a comprehensive bone phenotype analysis of mice with functional ablation of Fgf20. Methods The study conducts an extensive analysis of Fgf20 knockout mice compared to controls, incorporating microCT scanning, volumetric analysis, Fgf9 subfamily expression and stimulation experiment and histological evaluation. Results The bone phenotype could be detected especially in the area of the lumbar and caudal part of the spine and in fingers. Regarding the spine, Fgf20-/- mice exhibited adhesions of the transverse process of the sixth lumbar vertebra to the pelvis as well as malformations in the distal part of their tails. Preaxial polydactyly and polysyndactyly in varying degrees of severity were also detected. High resolution microCT analysis of distal femurs and the fourth lumbar vertebra showed significant differences in structure and mineralization in both cortical and trabecular bone. These findings were histologically validated and may be associated with the expression of Fgf20 in chondrocytes and their progenitors. Moreover, histological sections demonstrated increased bone tissue formation, disruption of Fgf20-/- femur cartilage, and cellular-level alterations, particularly in osteoclasts. We also observed molar dysmorphology, including root taurodontism, and described variations in mineralization and dentin thickness. Discussion Our analysis provides evidence that Fgf20, together with other members of the Fgf9 subfamily, plays a crucial regulatory role in skeletal development and bone homeostasis.
Collapse
Affiliation(s)
- Sylvie Dlugosova
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czechia
| | - Frantisek Spoutil
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czechia
| | | | - Betul Melike Ogan
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czechia
| | - Michaela Prochazkova
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czechia
| | - Olha Fedosieieva
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czechia
| | - Petr Nickl
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czechia
| | - Goretti Aranaz Novaliches
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czechia
| | - Radislav Sedlacek
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czechia
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czechia
| | - Jan Prochazka
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czechia
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czechia
| |
Collapse
|
4
|
Muksuris K, Scarisbrick DM, Mahoney JJ, Cherkasova MV. Noninvasive Neuromodulation in Parkinson's Disease: Insights from Animal Models. J Clin Med 2023; 12:5448. [PMID: 37685514 PMCID: PMC10487610 DOI: 10.3390/jcm12175448] [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: 07/17/2023] [Revised: 08/14/2023] [Accepted: 08/17/2023] [Indexed: 09/10/2023] Open
Abstract
The mainstay treatments for Parkinson's Disease (PD) have been limited to pharmacotherapy and deep brain stimulation. While these interventions are helpful, a new wave of research is investigating noninvasive neuromodulation methods as potential treatments. Some promising avenues have included transcranial magnetic stimulation (TMS), transcranial direct current stimulation (tDCS), electroconvulsive therapy (ECT), and focused ultrasound (FUS). While these methods are being tested in PD patients, investigations in animal models of PD have sought to elucidate their therapeutic mechanisms. In this rapid review, we assess the available animal literature on these noninvasive techniques and discuss the possible mechanisms mediating their therapeutic effects based on these findings.
Collapse
Affiliation(s)
- Katherine Muksuris
- Department of Psychology, West Virginia University, Morgantown, WV 26506, USA
| | - David M. Scarisbrick
- Department of Behavioral Medicine and Psychiatry, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| | - James J. Mahoney
- Department of Behavioral Medicine and Psychiatry, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Mariya V. Cherkasova
- Department of Psychology, West Virginia University, Morgantown, WV 26506, USA
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| |
Collapse
|
5
|
Villadiego J, García-Swinburn R, García-González D, Lebrón-Galán R, Murcia-Belmonte V, García-Roldán E, Suárez-Luna N, Nombela C, Marchena M, de Castro F, Toledo-Aral JJ. Extracellular matrix protein anosmin-1 overexpression alters dopaminergic phenotype in the CNS and the PNS with no pathogenic consequences in a MPTP model of Parkinson's disease. Brain Struct Funct 2023; 228:907-920. [PMID: 36995433 PMCID: PMC10147818 DOI: 10.1007/s00429-023-02631-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 03/15/2023] [Indexed: 03/31/2023]
Abstract
The development and survival of dopaminergic neurons are influenced by the fibroblast growth factor (FGF) pathway. Anosmin-1 (A1) is an extracellular matrix protein that acts as a major regulator of this signaling pathway, controlling FGF diffusion, and receptor interaction and shuttling. In particular, previous work showed that A1 overexpression results in more dopaminergic neurons in the olfactory bulb. Prompted by those intriguing results, in this study, we investigated the effects of A1 overexpression on different populations of catecholaminergic neurons in the central (CNS) and the peripheral nervous systems (PNS). We found that A1 overexpression increases the number of dopaminergic substantia nigra pars compacta (SNpc) neurons and alters the striosome/matrix organization of the striatum. Interestingly, these numerical and morphological changes in the nigrostriatal pathway of A1-mice did not confer an altered susceptibility to experimental MPTP-parkinsonism with respect to wild-type controls. Moreover, the study of the effects of A1 overexpression was extended to different dopaminergic tissues associated with the PNS, detecting a significant reduction in the number of dopaminergic chemosensitive carotid body glomus cells in A1-mice. Overall, our work shows that A1 regulates the development and survival of dopaminergic neurons in different nuclei of the mammalian nervous system.
Collapse
Affiliation(s)
- Javier Villadiego
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío-CSIC-Universidad de Sevilla, Avda. Manuel Siurot s/n, 41013, Seville, Spain
- Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, 41009, Sevilla, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Roberto García-Swinburn
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío-CSIC-Universidad de Sevilla, Avda. Manuel Siurot s/n, 41013, Seville, Spain
- Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, 41009, Sevilla, Spain
| | - Diego García-González
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío-CSIC-Universidad de Sevilla, Avda. Manuel Siurot s/n, 41013, Seville, Spain
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos, 45071, Toledo, Spain
| | - Rafael Lebrón-Galán
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos, 45071, Toledo, Spain
| | - Verónica Murcia-Belmonte
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos, 45071, Toledo, Spain
- Instituto de Neurociencias, UMH-CSIC, Sant Joan d´Alacant, 03550, Alicante, Spain
| | - Ernesto García-Roldán
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío-CSIC-Universidad de Sevilla, Avda. Manuel Siurot s/n, 41013, Seville, Spain
- Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, 41009, Sevilla, Spain
- Servicio de Neurología y Neurofisiología Clínica, Hospital Universitario Virgen del Rocío, 41013, Sevilla, Spain
| | - Nela Suárez-Luna
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío-CSIC-Universidad de Sevilla, Avda. Manuel Siurot s/n, 41013, Seville, Spain
- Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, 41009, Sevilla, Spain
| | - Cristina Nombela
- Departamento de Psicología Biológica y de la Salud, Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | - Miguel Marchena
- Grupo de Neurobiología del Desarrollo-GNDe, Instituto Cajal-CSIC, Avenida Doctor Arce 37, 28002, Madrid, Spain
- Departamento de Medicina, Universidad Europea de Madrid-UEM, Villaviciosa de Odón, 28670, Madrid, Spain
| | - Fernando de Castro
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos, 45071, Toledo, Spain.
- Grupo de Neurobiología del Desarrollo-GNDe, Instituto Cajal-CSIC, Avenida Doctor Arce 37, 28002, Madrid, Spain.
| | - Juan José Toledo-Aral
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío-CSIC-Universidad de Sevilla, Avda. Manuel Siurot s/n, 41013, Seville, Spain.
- Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, 41009, Sevilla, Spain.
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| |
Collapse
|
6
|
Huang Q, Chen C, Chen W, Cai C, Xing H, Li J, Li M, Ma S. Cell type- and region-specific translatomes in an MPTP mouse model of Parkinson's disease. Neurobiol Dis 2023; 180:106105. [PMID: 36977454 DOI: 10.1016/j.nbd.2023.106105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/16/2023] [Accepted: 03/24/2023] [Indexed: 03/28/2023] Open
Abstract
Parkinson's disease (PD) is the most common neurodegenerative movement disorder, characterized by the progressive loss of nigrostriatal dopaminergic neurons (DANs), involving the dysregulation of both neurons and glial cells. Cell type- and region-specific gene expression profiles can provide an effective source for revealing the mechanisms of PD. In this study, we adopted the RiboTag approach to obtain cell type (DAN, microglia, astrocytes)- and brain region (substantia nigra, caudate-putamen)-specific translatomes at an early stage in an MPTP-induced mouse model of PD. Through DAN-specific translatome analysis, the glycosphingolipid biosynthetic process was identified as a significantly downregulated pathway in the MPTP-treated mice. ST8Sia6, a key downregulated gene related to glycosphingolipid biosynthesis, was confirmed to be downregulated in nigral DANs from postmortem brains of patients with PD. Specific expression of ST8Sia6 in DANs exerts anti-inflammatory and neuroprotective effects in MPTP-treated mice. Through cell type (microglia vs. astrocyte) and brain region (substantia nigra vs. caudate-putamen) comparisons, nigral microglia showed the most intense immune responses. Microglia and astrocytes in the substantia nigra showed similar levels of activation in interferon-related pathways and interferon gamma (IFNG) was identified as the top upstream regulator in both cell types. This work highlights that the glycosphingolipid metabolism pathway in the DAN is involved in neuroinflammation and neurodegeneration in an MPTP mouse model of PD and provides a new data source for elucidating the pathogenesis of PD.
Collapse
|
7
|
Huang W, Qiu W, Chen K, Ye S, Wang D, Hu J, Xu H, Lin L, Li X. Research progress of fibroblast growth factor in nervous system diseases. Zhejiang Da Xue Xue Bao Yi Xue Ban 2022; 51:738-749. [PMID: 36915973 PMCID: PMC10262007 DOI: 10.3724/zdxbyxb-2022-0180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 08/25/2022] [Indexed: 06/17/2023]
Abstract
Fibroblast growth factors (FGF) are a group of structurally related polypeptides which constitute an elaborate signaling system with their receptors. Evidence accumulated in the years suggests that the FGF family plays a key role in the repair of central nervous system injury. The main protective mechanisms include activating the expression of PI3K-Akt, peroxisome proliferator-activated receptor (PPARγ) and other signals; inhibiting NF-κB-mediated inflammatory response, oxidative stress and apoptosis; regulating neuronal differentiation and neuronal excitability as well as participating in protection of neurovascular units and nerve function repair. This paper comprehensively summarizes the latest research progress in FGF signaling related to diseases of the central nervous system such as cerebral infarction, cerebral hemorrhage, traumatic brain injury, Alzheimer's disease, Parkinson's disease, epilepsy and depression, aiming to provide scientific basis and reference for the development of innovative FGF drugs for the prevention and treatment of neurological diseases.
Collapse
Affiliation(s)
- Wenting Huang
- 1. Department of Neurology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, China
| | - Wanhua Qiu
- 2. School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Kun Chen
- 2. School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Shasha Ye
- 2. School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Dongxue Wang
- 2. School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Jian Hu
- 2. School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Huiqin Xu
- 1. Department of Neurology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, China
| | - Li Lin
- 2. School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Xiaokun Li
- 2. School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| |
Collapse
|
8
|
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
|
9
|
Li J, Yi M, Li B, Yin S, Zhang Y, Huang Z, Shu L, Zhang Y. Polymorphism of neurodegeneration-related genes associated with Parkinson’s disease risk. Neurol Sci 2022; 43:5301-5312. [DOI: 10.1007/s10072-022-06192-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 04/29/2022] [Indexed: 12/23/2022]
|
10
|
Roy B, Lee E, Li T, Rampersaud M. Role of miRNAs in Neurodegeneration: From Disease Cause to Tools of Biomarker Discovery and Therapeutics. Genes (Basel) 2022; 13:genes13030425. [PMID: 35327979 PMCID: PMC8951370 DOI: 10.3390/genes13030425] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/14/2022] [Accepted: 02/18/2022] [Indexed: 11/16/2022] Open
Abstract
Neurodegenerative diseases originate from neuronal loss in the central nervous system (CNS). These debilitating diseases progress with age and have become common due to an increase in longevity. The National Institute of Environmental Health Science’s 2021 annual report suggests around 6.2 million Americans are living with Alzheimer’s disease, and there is a possibility that there will be 1.2 million Parkinson’s disease patients in the USA by 2030. There is no clear-cut universal mechanism for identifying neurodegenerative diseases, and therefore, they pose a challenge for neurobiology scientists. Genetic and environmental factors modulate these diseases leading to familial or sporadic forms. Prior studies have shown that miRNA levels are altered during the course of the disease, thereby suggesting that these noncoding RNAs may be the contributing factor in neurodegeneration. In this review, we highlight the role of miRNAs in the pathogenesis of neurodegenerative diseases. Through this review, we aim to achieve four main objectives: First, we highlight how dysregulation of miRNA biogenesis led to these diseases. Second, we highlight the computational or bioinformatics tools required to identify the putative molecular targets of miRNAs, leading to biological molecular pathways or mechanisms involved in these diseases. Third, we focus on the dysregulation of miRNAs and their target genes leading to several neurodegenerative diseases. In the final section, we highlight the use of miRNAs as potential diagnostic biomarkers in the early asymptomatic preclinical diagnosis of these age-dependent debilitating diseases. Additionally, we discuss the challenges and advances in the development of miRNA therapeutics for brain targeting. We list some of the innovative strategies employed to deliver miRNA into target cells and the relevance of these viral and non-viral carrier systems in RNA therapy for neurodegenerative diseases. In summary, this review highlights the relevance of studying brain-enriched miRNAs, the mechanisms underlying their regulation of target gene expression, their dysregulation leading to progressive neurodegeneration, and their potential for biomarker marker and therapeutic intervention. This review thereby highlights ways for the effective diagnosis and prevention of these neurodegenerative disorders in the near future.
Collapse
Affiliation(s)
- Bidisha Roy
- Life Science Centre, Department of Biological Sciences, Rutgers University-Newark, Newark, NJ 07012, USA
- Correspondence:
| | - Erica Lee
- Department of Pathology, Icahn School of Medicine, New York, NY 10029, USA; (E.L.); (T.L.); (M.R.)
| | - Teresa Li
- Department of Pathology, Icahn School of Medicine, New York, NY 10029, USA; (E.L.); (T.L.); (M.R.)
| | - Maria Rampersaud
- Department of Pathology, Icahn School of Medicine, New York, NY 10029, USA; (E.L.); (T.L.); (M.R.)
| |
Collapse
|
11
|
Quan W, Li J, Liu L, Zhang Q, Qin Y, Pei X, Chen J. Quantitative assessment of the effect of FGF20 rs1721100 and rs12720208 variant on the risk of sporadic Parkinson's disease: a meta-analysis. Neurol Sci 2021; 43:3145-3152. [PMID: 34845561 DOI: 10.1007/s10072-021-05754-6] [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: 06/23/2021] [Accepted: 11/12/2021] [Indexed: 11/30/2022]
Abstract
OBJECTIVES While many studies have investigated the associations between fibroblast growth factor 20 (FGF20) rs1721100 (C/G) and rs12720208 (C/T) polymorphisms and susceptibility to Parkinson's disease (PD), their results are controversial. Our present meta-analysis estimated the overall association between FGF20 rs1721100 and rs12720208 polymorphisms and the risk of sporadic PD. METHODS We performed a comprehensive literature search of the PubMed, Web of Science, Embase, Chinese National Knowledge Infrastructure, and Wanfang Medicine electronic databases, which was updated in April 2021. Based on strict inclusion and exclusion criteria, the analysis included a total of 10 papers involving 14 studies with 5262 cases of PD and 6075 controls. Review Manager 5.4 software was used to assess the available data from each study. The pooled odds ratios (ORs) and 95% confidence intervals (CIs) were calculated to assess the association between the FGF20 rs1721100 and rs12720208 polymorphisms and sporadic PD risk. RESULTS Our results showed that the FGF20 rs1721100 G allele frequency and genotype distribution did not differ between PD patients and controls. Similarly, the FGF20 rs12720208 T allele frequency and genotype distribution did not differ significantly between the two groups. A subgroup analysis of Asian and Caucasian populations also showed the same results. CONCLUSIONS The results of this meta-analysis indicated that neither the rs1721100 C/G nor the rs12720208 C/T variants were associated with sporadic PD susceptibility.
Collapse
Affiliation(s)
- Wei Quan
- Department of Neurology, China-Japan Union Hospital of Jilin University, No. 126, Xian Tai Road, Changchun, 130000, Jilin, China
| | - Jia Li
- Department of Neurology, China-Japan Union Hospital of Jilin University, No. 126, Xian Tai Road, Changchun, 130000, Jilin, China
| | - Li Liu
- Department of Neurology, China-Japan Union Hospital of Jilin University, No. 126, Xian Tai Road, Changchun, 130000, Jilin, China
| | - Qinghui Zhang
- Department of Neurology, China-Japan Union Hospital of Jilin University, No. 126, Xian Tai Road, Changchun, 130000, Jilin, China
| | - Yidan Qin
- Department of Neurology, China-Japan Union Hospital of Jilin University, No. 126, Xian Tai Road, Changchun, 130000, Jilin, China
| | - Xiaochen Pei
- Department of Neurology, China-Japan Union Hospital of Jilin University, No. 126, Xian Tai Road, Changchun, 130000, Jilin, China
| | - Jiajun Chen
- Department of Neurology, China-Japan Union Hospital of Jilin University, No. 126, Xian Tai Road, Changchun, 130000, Jilin, China.
| |
Collapse
|
12
|
Pu JL, Lin ZH, Zheng R, Yan YQ, Xue NJ, Yin XZ, Zhang BR. Association analysis of SYT11, FGF20, GCH1 rare variants in Parkinson's disease. CNS Neurosci Ther 2021; 28:175-177. [PMID: 34674384 PMCID: PMC8673698 DOI: 10.1111/cns.13745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 01/28/2023] Open
Affiliation(s)
- Jia-Li Pu
- Department of Neurology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Zhi-Hao Lin
- Department of Neurology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ran Zheng
- Department of Neurology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yi-Qun Yan
- Department of Neurology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Nai-Jia Xue
- Department of Neurology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xin-Zhen Yin
- Department of Neurology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Bao-Rong Zhang
- Department of Neurology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
13
|
Campbell RR, Chen S, Beardwood JH, López AJ, Pham LV, Keiser AM, Childs JE, Matheos DP, Swarup V, Baldi P, Wood MA. Cocaine induces paradigm-specific changes to the transcriptome within the ventral tegmental area. Neuropsychopharmacology 2021; 46:1768-1779. [PMID: 34155331 PMCID: PMC8357835 DOI: 10.1038/s41386-021-01031-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 04/29/2021] [Accepted: 04/29/2021] [Indexed: 12/16/2022]
Abstract
During the initial stages of drug use, cocaine-induced neuroadaptations within the ventral tegmental area (VTA) are critical for drug-associated cue learning and drug reinforcement processes. These neuroadaptations occur, in part, from alterations to the transcriptome. Although cocaine-induced transcriptional mechanisms within the VTA have been examined, various regimens and paradigms have been employed to examine candidate target genes. In order to identify key genes and biological processes regulating cocaine-induced processes, we employed genome-wide RNA-sequencing to analyze transcriptional profiles within the VTA from male mice that underwent one of four commonly used paradigms: acute home cage injections of cocaine, chronic home cage injections of cocaine, cocaine-conditioning, or intravenous-self administration of cocaine. We found that cocaine alters distinct sets of VTA genes within each exposure paradigm. Using behavioral measures from cocaine self-administering mice, we also found several genes whose expression patterns corelate with cocaine intake. In addition to overall gene expression levels, we identified several predicted upstream regulators of cocaine-induced transcription shared across all paradigms. Although distinct gene sets were altered across cocaine exposure paradigms, we found, from Gene Ontology (GO) term analysis, that biological processes important for energy regulation and synaptic plasticity were affected across all cocaine paradigms. Coexpression analysis also identified gene networks that are altered by cocaine. These data indicate that cocaine alters networks enriched with glial cell markers of the VTA that are involved in gene regulation and synaptic processes. Our analyses demonstrate that transcriptional changes within the VTA depend on the route, dose and context of cocaine exposure, and highlight several biological processes affected by cocaine. Overall, these findings provide a unique resource of gene expression data for future studies examining novel cocaine gene targets that regulate drug-associated behaviors.
Collapse
Affiliation(s)
- Rianne R Campbell
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine, CA, USA
- UC Irvine Center for Addiction Neuroscience, School of Biological Sciences, University of California, Irvine, CA, USA
- Center for the Neurobiology of Learning and Memory, School of Biological Sciences, University of California, Irvine, CA, USA
| | - Siwei Chen
- Department of Computer Science, University of California, Irvine, CA, USA
- Institute for Genomics and Bioinformatics, University of California, Irvine, CA, USA
| | - Joy H Beardwood
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine, CA, USA
- UC Irvine Center for Addiction Neuroscience, School of Biological Sciences, University of California, Irvine, CA, USA
- Center for the Neurobiology of Learning and Memory, School of Biological Sciences, University of California, Irvine, CA, USA
| | - Alberto J López
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Lilyana V Pham
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine, CA, USA
- UC Irvine Center for Addiction Neuroscience, School of Biological Sciences, University of California, Irvine, CA, USA
- Center for the Neurobiology of Learning and Memory, School of Biological Sciences, University of California, Irvine, CA, USA
| | - Ashley M Keiser
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine, CA, USA
- Center for the Neurobiology of Learning and Memory, School of Biological Sciences, University of California, Irvine, CA, USA
| | - Jessica E Childs
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine, CA, USA
- UC Irvine Center for Addiction Neuroscience, School of Biological Sciences, University of California, Irvine, CA, USA
- Center for the Neurobiology of Learning and Memory, School of Biological Sciences, University of California, Irvine, CA, USA
| | - Dina P Matheos
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine, CA, USA
- UC Irvine Center for Addiction Neuroscience, School of Biological Sciences, University of California, Irvine, CA, USA
- Center for the Neurobiology of Learning and Memory, School of Biological Sciences, University of California, Irvine, CA, USA
| | - Vivek Swarup
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine, CA, USA
| | - Pierre Baldi
- Center for the Neurobiology of Learning and Memory, School of Biological Sciences, University of California, Irvine, CA, USA
- Department of Computer Science, University of California, Irvine, CA, USA
- Institute for Genomics and Bioinformatics, University of California, Irvine, CA, USA
| | - Marcelo A Wood
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine, CA, USA.
- UC Irvine Center for Addiction Neuroscience, School of Biological Sciences, University of California, Irvine, CA, USA.
- Center for the Neurobiology of Learning and Memory, School of Biological Sciences, University of California, Irvine, CA, USA.
| |
Collapse
|
14
|
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: 65] [Impact Index Per Article: 16.3] [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
|
15
|
Liu Y, Deng J, Liu Y, Li W, Nie X. FGF, Mechanism of Action, Role in Parkinson's Disease, and Therapeutics. Front Pharmacol 2021; 12:675725. [PMID: 34234672 PMCID: PMC8255968 DOI: 10.3389/fphar.2021.675725] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 06/09/2021] [Indexed: 12/17/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disease associated with severe disability and adverse effects on life quality. In PD, motor dysfunction can occur, such as quiescence, muscle stiffness, and postural instability. PD is also associated with autonomic nervous dysfunction, sleep disorders, psychiatric symptoms, and other non-motor symptoms. Degeneration of dopaminergic neurons in the substantia nigra compact (SNPC), Lewy body, and neuroinflammation are the main pathological features of PD. The death or dysfunction of dopaminergic neurons in the dense part of the substantia nigra leads to dopamine deficiency in the basal ganglia and motor dysfunction. The formation of the Lewy body is associated with the misfolding of α-synuclein, which becomes insoluble and abnormally aggregated. Astrocytes and microglia mainly cause neuroinflammation, and the activation of a variety of pro-inflammatory transcription factors and regulatory proteins leads to the degeneration of dopaminergic neurons. At present, PD is mainly treated with drugs that increase dopamine concentration or directly stimulate dopamine receptors. Fibroblast growth factor (FGF) is a family of cellular signaling proteins strongly associated with neurodegenerative diseases such as PD. FGF and its receptor (FGFR) play an essential role in the development and maintenance of the nervous system as well as in neuroinflammation and have been shown to improve the survival rate of dopaminergic neurons. This paper summarized the mechanism of FGF and its receptors in the pathological process of PD and related signaling pathways, involving the development and protection of dopaminergic neurons in SNPC, α-synuclein aggregation, mitochondrial dysfunction, and neuroinflammation. It provides a reference for developing drugs to slow down or prevent the potential of PD.
Collapse
Affiliation(s)
- Yiqiu Liu
- College of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Junyu Deng
- College of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Ye Liu
- College of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Wei Li
- College of Pharmacy, Zunyi Medical University, Zunyi, China
- Joint International Research Laboratory of Ethnomedicine of Chinese Ministry of Education, College of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Xuqiang Nie
- College of Pharmacy, Zunyi Medical University, Zunyi, China
- Joint International Research Laboratory of Ethnomedicine of Chinese Ministry of Education, College of Pharmacy, Zunyi Medical University, Zunyi, China
- Key Lab of the Basic Pharmacology of the Ministry of Education, College of Pharmacy, Zunyi Medical University, Zunyi, China
| |
Collapse
|
16
|
Seitz T, Hellerbrand C. Role of fibroblast growth factor signalling in hepatic fibrosis. Liver Int 2021; 41:1201-1215. [PMID: 33655624 DOI: 10.1111/liv.14863] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/22/2021] [Accepted: 02/25/2021] [Indexed: 12/11/2022]
Abstract
Fibrotic remodelling is a highly conserved protective response to tissue injury and it is essential for the maintenance of structural and functional tissue integrity. Also hepatic fibrosis can be considered as a wound-healing response to liver injury, reflecting a balance between liver repair and scar formation. In contrast, pathological fibrosis corresponds to impaired wound healing. Usually, the liver regenerates after acute injury. However, if the damaging mechanisms persist, the liver reacts with progressive and uncontrolled accumulation of extracellular matrix proteins. Eventually, excessive fibrosis can lead to cirrhosis and hepatic failure. Furthermore, cirrhosis is the major risk factor for the development of hepatocellular cancer (HCC). Therefore, hepatic fibrosis is the most critical pathological factor that determines the morbidity and mortality of patients with chronic liver disease. Still, no effective anti-fibrogenic therapies exist, despite the very high medical need. The regulation of fibroblast growth factor (FGF) signalling is a prerequisite for adequate wound healing, repair and homeostasis in various tissues and organs. The FGF family comprises 22 proteins that can be classified into paracrine, intracrine and endocrine factors. Most FGFs signal through transmembrane tyrosine kinase FGF receptors (FGFRs). Although FGFRs are promising targets for the treatment of HCC, the expression and function of FGFR-ligands in hepatic fibrosis is still poorly understood. This review summarizes the latest advances in our understanding of FGF signalling in hepatic fibrosis. Furthermore, the potential of FGFs as targets for the treatment of hepatic fibrosis and remaining challenges for the field are discussed.
Collapse
Affiliation(s)
- Tatjana Seitz
- Institute of Biochemistry, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Claus Hellerbrand
- Institute of Biochemistry, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
17
|
Chiang HL, Wu YR, Chen YC, Fung HC, Chen CM. Fibroblast Growth Factor 20 Gene Polymorphism in Parkinson's Disease in Asian Population: A Meta-Analysis. Genes (Basel) 2021; 12:genes12050674. [PMID: 33947140 PMCID: PMC8145858 DOI: 10.3390/genes12050674] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 04/25/2021] [Accepted: 04/26/2021] [Indexed: 11/16/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disease with the pathological hallmark of Lewy bodies and Lewy neurites composed of α-synuclein. The SNP rs591323 is one of the risk loci located near the FGF20 gene that has been implicated in PD. The variation of FGF20 in the 3' untranslated region was shown to increase α-synuclein expression. We examined the association of rs591323 with the risk of PD in a Taiwanese population and conducted a meta-analysis, including our study and two other studies from China, to further confirm the role of this SNP in Taiwanese/Chinese populations. A total of 586 patients with PD and 586 health controls (HCs) were included in our study. We found that the minor allele (A) and the AA + GA genotype under the dominant model are significantly less frequent in PD than in controls. The meta-analysis consisted of 1950 patients with PD and 2073 healthy controls from three studies. There was significant association between rs591323 and the risk of PD in the additive (Z = -3.96; p < 0.0001) and the dominant models (Z = -4.01; p < 0.0001). Our study results and the meta-analysis support the possible protective role of the rs591323 A allele in PD in Taiwanese/Chinese populations.
Collapse
Affiliation(s)
- Han-Lin Chiang
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei 112, Taiwan;
| | - Yih-Ru Wu
- Chang Gung Memorial Hospital Linkou Medical Center and College of Medicine, Department of Neurology, Chang Gung University, Taoyuan 333, Taiwan; (Y.-R.W.); (Y.-C.C.)
| | - Yi-Chun Chen
- Chang Gung Memorial Hospital Linkou Medical Center and College of Medicine, Department of Neurology, Chang Gung University, Taoyuan 333, Taiwan; (Y.-R.W.); (Y.-C.C.)
| | - Hon-Chung Fung
- Fu Jen Faculty of Theology of St. Robert Bellarmine, Fu Jen University Clinic, Taipei 242, Taiwan;
| | - Chiung-Mei Chen
- Chang Gung Memorial Hospital Linkou Medical Center and College of Medicine, Department of Neurology, Chang Gung University, Taoyuan 333, Taiwan; (Y.-R.W.); (Y.-C.C.)
- Correspondence: ; Tel.: +886-3-3281200 (ext. 8729)
| |
Collapse
|
18
|
Rudakou U, Yu E, Krohn L, Ruskey JA, Asayesh F, Dauvilliers Y, Spiegelman D, Greenbaum L, Fahn S, Waters CH, Dupré N, Rouleau GA, Hassin-Baer S, Fon EA, Alcalay RN, Gan-Or Z. Targeted sequencing of Parkinson's disease loci genes highlights SYT11, FGF20 and other associations. Brain 2021; 144:462-472. [PMID: 33349842 DOI: 10.1093/brain/awaa401] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/13/2020] [Accepted: 09/09/2020] [Indexed: 01/13/2023] Open
Abstract
Genome-wide association studies (GWAS) have identified numerous loci associated with Parkinson's disease. The specific genes and variants that drive the associations within the vast majority of these loci are unknown. We aimed to perform a comprehensive analysis of selected genes to determine the potential role of rare and common genetic variants within these loci. We fully sequenced 32 genes from 25 loci previously associated with Parkinson's disease in 2657 patients and 3647 controls from three cohorts. Capture was done using molecular inversion probes targeting the exons, exon-intron boundaries and untranslated regions (UTRs) of the genes of interest, followed by sequencing. Quality control was performed to include only high-quality variants. We examined the role of rare variants (minor allele frequency < 0.01) using optimized sequence Kernel association tests. The association of common variants was estimated using regression models adjusted for age, sex and ethnicity as required in each cohort, followed by a meta-analysis. After Bonferroni correction, we identified a burden of rare variants in SYT11, FGF20 and GCH1 associated with Parkinson's disease. Nominal associations were identified in 21 additional genes. Previous reports suggested that the SYT11 GWAS association is driven by variants in the nearby GBA gene. However, the association of SYT11 was mainly driven by a rare 3' UTR variant (rs945006601) and was independent of GBA variants (P = 5.23 × 10-5 after exclusion of all GBA variant carriers). The association of FGF20 was driven by a rare 5' UTR variant (rs1034608171) located in the promoter region. The previously reported association of GCH1 with Parkinson's disease is driven by rare non-synonymous variants, some of which are known to cause dopamine-responsive dystonia. We also identified two LRRK2 variants, p.Arg793Met and p.Gln1353Lys, in 10 and eight controls, respectively, but not in patients. We identified common variants associated with Parkinson's disease in MAPT, TMEM175, BST1, SNCA and GPNMB, which are all in strong linkage disequilibrium with known GWAS hits in their respective loci. A common coding PM20D1 variant, p.Ile149Val, was nominally associated with reduced risk of Parkinson's disease (odds ratio 0.73, 95% confidence interval 0.60-0.89, P = 1.161 × 10-3). This variant is not in linkage disequilibrium with the top GWAS hits within this locus and may represent a novel association. These results further demonstrate the importance of fine mapping of GWAS loci, and suggest that SYT11, FGF20, and potentially PM20D1, BST1 and GPNMB should be considered for future studies as possible Parkinson's disease-related genes.
Collapse
Affiliation(s)
- Uladzislau Rudakou
- Department of Human Genetics, McGill University, Montréal, QC, H3A 1A1, Canada.,Montreal Neurological Institute, McGill University, Montréal, QC, H3A 1A1, Canada
| | - Eric Yu
- Department of Human Genetics, McGill University, Montréal, QC, H3A 1A1, Canada.,Montreal Neurological Institute, McGill University, Montréal, QC, H3A 1A1, Canada
| | - Lynne Krohn
- Department of Human Genetics, McGill University, Montréal, QC, H3A 1A1, Canada.,Montreal Neurological Institute, McGill University, Montréal, QC, H3A 1A1, Canada
| | - Jennifer A Ruskey
- Montreal Neurological Institute, McGill University, Montréal, QC, H3A 1A1, Canada.,Department of Neurology and Neurosurgery, McGill University, Montréal, QC, H3A 1A1, Canada
| | - Farnaz Asayesh
- Montreal Neurological Institute, McGill University, Montréal, QC, H3A 1A1, Canada.,Department of Neurology and Neurosurgery, McGill University, Montréal, QC, H3A 1A1, Canada
| | - Yves Dauvilliers
- National Reference Center for Narcolepsy, Sleep Unit, Department of Neurology, Gui-de-Chauliac Hospital, CHU Montpellier, University of Montpellier, Inserm U1061, Montpellier, France
| | - Dan Spiegelman
- Montreal Neurological Institute, McGill University, Montréal, QC, H3A 1A1, Canada.,Department of Neurology and Neurosurgery, McGill University, Montréal, QC, H3A 1A1, Canada
| | - Lior Greenbaum
- The Danek Gertner Institute of Human Genetics, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel.,The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Stanley Fahn
- Department of Neurology, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY 10032, USA
| | - Cheryl H Waters
- Department of Neurology, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY 10032, USA
| | - Nicolas Dupré
- Division of Neurosciences, CHU de Québec, Université Laval, Québec City, QC, G1V 0A6, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Québec City, QC, G1V 0A6, Canada
| | - Guy A Rouleau
- Department of Human Genetics, McGill University, Montréal, QC, H3A 1A1, Canada.,Montreal Neurological Institute, McGill University, Montréal, QC, H3A 1A1, Canada.,Department of Neurology and Neurosurgery, McGill University, Montréal, QC, H3A 1A1, Canada
| | - Sharon Hassin-Baer
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Neurology, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel.,Movement Disorders Institute, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Edward A Fon
- Montreal Neurological Institute, McGill University, Montréal, QC, H3A 1A1, Canada.,Department of Neurology and Neurosurgery, McGill University, Montréal, QC, H3A 1A1, Canada
| | - Roy N Alcalay
- Department of Neurology, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY 10032, USA.,Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY 10032, USA
| | - Ziv Gan-Or
- Department of Human Genetics, McGill University, Montréal, QC, H3A 1A1, Canada.,Montreal Neurological Institute, McGill University, Montréal, QC, H3A 1A1, Canada.,Department of Neurology and Neurosurgery, McGill University, Montréal, QC, H3A 1A1, Canada
| |
Collapse
|
19
|
Aloizou AM, Siokas V, Sapouni EM, Sita N, Liampas I, Brotis AG, Rakitskii VN, Burykina TI, Aschner M, Bogdanos DP, Tsatsakis A, Hadjigeorgiou GM, Dardiotis E. Parkinson's disease and pesticides: Are microRNAs the missing link? THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 744:140591. [PMID: 32721662 DOI: 10.1016/j.scitotenv.2020.140591] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/19/2020] [Accepted: 06/26/2020] [Indexed: 06/11/2023]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder that leads to significant morbidity and decline in the quality of life. It develops due to loss of dopaminergic neurons in the substantia nigra pars compacta, and among its pathogenic factors oxidative stress plays a critical role in disease progression. Pesticides are a broad class of chemicals widely used in agriculture and households for the protection of crops from insects and fungi. Several of them have been incriminated as risk factors for PD, but the underlying mechanisms have yet to be fully understood. MicroRNAs (miRNAs) are small, non-coding RNA molecules that play an important role in regulating mRNA translation and protein synthesis. miRNA levels have been shown to be affected in several diseases as well. Since the studies on the association between pesticides and PD have yet to reach definitive conclusions, here we review recent evidence on deregulated microRNAs upon pesticide exposure, and attempt to find an overlap between miRNAs deregulated in PD and pesticides, as a missing link between the two, and enhance future research in this direction.
Collapse
Affiliation(s)
- Athina-Maria Aloizou
- Department of Neurology, Laboratory of Neurogenetics, University Hospital of Larissa, Greece, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Vasileios Siokas
- Department of Neurology, Laboratory of Neurogenetics, University Hospital of Larissa, Greece, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece.
| | - Efstathia-Maria Sapouni
- Department of Neurology, Laboratory of Neurogenetics, University Hospital of Larissa, Greece, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Nikoleta Sita
- Department of Neurology, Laboratory of Neurogenetics, University Hospital of Larissa, Greece, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Ioannis Liampas
- Department of Neurology, Laboratory of Neurogenetics, University Hospital of Larissa, Greece, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Alexandros G Brotis
- Department of Neurosurgery, School of Medicine, University Hospital of Larissa, University of Thessaly, Larissa, Greece
| | - Valerii N Rakitskii
- The Federal Budgetary Establishment of Science "Federal Scientific Center of Hygiene named after F. F. Erisman" of the Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing, 2 Semashko street, Mytishchi, Moscow Oblast' 141014, Russian Federation
| | - Tatyana I Burykina
- Department of Analytical and Forensic Medical Toxicology, Sechenov University, 119048 Moscow, Russian Federation
| | - Michael Aschner
- Albert Einstein College of Medicine, Bronx, NY, USA; IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Dimitrios P Bogdanos
- Department of Rheumatology and clinical Immunology, University General Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, Viopolis 40500, Larissa, Greece
| | - Aristidis Tsatsakis
- The Federal Budgetary Establishment of Science "Federal Scientific Center of Hygiene named after F. F. Erisman" of the Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing, 2 Semashko street, Mytishchi, Moscow Oblast' 141014, Russian Federation; Department of Analytical and Forensic Medical Toxicology, Sechenov University, 119048 Moscow, Russian Federation; Laboratory of Toxicology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Georgios M Hadjigeorgiou
- Department of Neurology, Laboratory of Neurogenetics, University Hospital of Larissa, Greece, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece; Department of Neurology, Medical School, University of Cyprus, Nicosia, Cyprus
| | - Efthimios Dardiotis
- Department of Neurology, Laboratory of Neurogenetics, University Hospital of Larissa, Greece, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| |
Collapse
|
20
|
Park M, Kwon CH, Ha HK, Han M, Song SH. RNA-Seq identifies condition-specific biological signatures of ischemia-reperfusion injury in the human kidney. BMC Nephrol 2020; 21:398. [PMID: 32977749 PMCID: PMC7517631 DOI: 10.1186/s12882-020-02025-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 08/17/2020] [Indexed: 12/13/2022] Open
Abstract
Background Acute kidney injury (AKI) is defined as a sudden event of kidney failure or kidney damage within a short period. Ischemia-reperfusion injury (IRI) is a critical factor associated with severe AKI and end-stage kidney disease (ESKD). However, the biological mechanisms underlying ischemia and reperfusion are incompletely understood, owing to the complexity of these pathophysiological processes. We aimed to investigate the key biological pathways individually affected by ischemia and reperfusion at the transcriptome level. Results We analyzed the steady-state gene expression pattern of human kidney tissues from normal (pre-ischemia), ischemia, and reperfusion conditions using RNA-sequencing. Conventional differential expression and self-organizing map (SOM) clustering analyses followed by pathway analysis were performed. Differential expression analysis revealed the metabolic pathways dysregulated in ischemia. Cellular assembly, development and migration, and immune response-related pathways were dysregulated in reperfusion. SOM clustering analysis highlighted the ischemia-mediated significant dysregulation in metabolism, apoptosis, and fibrosis-related pathways, while cell growth, migration, and immune response-related pathways were highly dysregulated by reperfusion after ischemia. The expression of pro-apoptotic genes and death receptors was downregulated during ischemia, indicating the existence of a protective mechanism against ischemic injury. Reperfusion induced alterations in the expression of the genes associated with immune response such as inflammasome and antigen representing genes. Further, the genes related to cell growth and migration, such as AKT, KRAS, and those related to Rho signaling, were downregulated, suggestive of injury responses during reperfusion. Semaphorin 4D and plexin B1 levels were also downregulated. Conclusions We show that specific biological pathways were distinctively involved in ischemia and reperfusion during IRI, indicating that condition-specific therapeutic strategies may be imperative to prevent severe kidney damage after IRI in the clinical setting.
Collapse
Affiliation(s)
- Meeyoung Park
- Biomedical Research Institute, Pusan National University Hospital, Busan, South Korea
| | - Chae Hwa Kwon
- Biomedical Research Institute, Pusan National University Hospital, Busan, South Korea
| | - Hong Koo Ha
- Biomedical Research Institute, Pusan National University Hospital, Busan, South Korea
| | - Miyeun Han
- Department of Urology, Pusan National University Hospital, Busan, South Korea
| | - Sang Heon Song
- Department of Internal Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan, South Korea.
| |
Collapse
|
21
|
MicroRNAs Dysregulation and Mitochondrial Dysfunction in Neurodegenerative Diseases. Int J Mol Sci 2020; 21:ijms21175986. [PMID: 32825273 PMCID: PMC7504116 DOI: 10.3390/ijms21175986] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/12/2020] [Accepted: 08/18/2020] [Indexed: 02/06/2023] Open
Abstract
Neurodegenerative diseases are debilitating and currently incurable conditions causing severe cognitive and motor impairments, defined by the progressive deterioration of neuronal structure and function, eventually causing neuronal loss. Understand the molecular and cellular mechanisms underlying these disorders are essential to develop therapeutic approaches. MicroRNAs (miRNAs) are short non-coding RNAs implicated in gene expression regulation at the post-transcriptional level. Moreover, miRNAs are crucial for different processes, including cell growth, signal transmission, apoptosis, cancer and aging-related neurodegenerative diseases. Altered miRNAs levels have been associated with the formation of reactive oxygen species (ROS) and mitochondrial dysfunction. Mitochondrial dysfunction and ROS formation occur in many neurodegenerative diseases such as Alzheimer's, Parkinson's and Huntington's diseases. The crosstalk existing among oxidative stress, mitochondrial dysfunction and miRNAs dysregulation plays a pivotal role in the onset and progression of neurodegenerative diseases. Based on this evidence, in this review, with a focus on miRNAs and their role in mitochondrial dysfunction in aging-related neurodegenerative diseases, with a focus on their potential as diagnostic biomarkers and therapeutic targets.
Collapse
|
22
|
Niu J, Xie J, Guo K, Zhang X, Xia F, Zhao X, Song L, Zhuge D, Li X, Zhao Y, Huang Z. Efficient treatment of Parkinson's disease using ultrasonography-guided rhFGF20 proteoliposomes. Drug Deliv 2018; 25:1560-1569. [PMID: 30043675 PMCID: PMC6060384 DOI: 10.1080/10717544.2018.1482972] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 05/25/2018] [Accepted: 05/28/2018] [Indexed: 12/11/2022] Open
Abstract
Fibroblast growth factor-20 (FGF20) is a paracrine member of the FGF family that is preferentially expressed in the substantia nigra pars compacta (SNpc). Previous studies have demonstrated that FGF20 enhances the survival of dopaminergic neurons suggesting the potential use of FGF20 to treat Parkinson's disease (PD). However, the reduced solubility of the bacterial recombinant human FGF20 (rhFGF20) and the absence of efficient strategies to transport rhFGF20 across the blood-brain barrier (BBB) have halted its clinical application. In the present study, we have examined the efficiency of fuzing a small ubiquitin-related modifier (SUMO) to rhFGF20 to enhance its soluble expression and further investigated the efficacy of FUS-guided, rhFGF20-liposome transport across the BBB. We also examined the bioavailability and behavioral improvement in a 6-hydroxydopamine-lesioned rat model of PD following 2 weeks' FUS-liposomal combinatorial treatment. Our results showed that, in contrast with rhFGF20 or LIP-FGF20, the FUS-LIP-rhFGF20 treatment could significantly improve the apomorphine-induced rotations by protecting against the loss of dopaminergic neurons in the SNpc. Our Results suggest that our combinatorial method would help overcome key challenges that hinder the currently available methods for the use of rhFGF20 in PD treatment.
Collapse
Affiliation(s)
- Jianlou Niu
- School of Pharmaceutical Sciences & Center for Structural Biology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Junjun Xie
- School of Pharmaceutical Sciences & Center for Structural Biology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Kaiwen Guo
- School of Pharmaceutical Sciences & Center for Structural Biology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaomin Zhang
- School of Pharmaceutical Sciences & Center for Structural Biology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Feng Xia
- School of Pharmaceutical Sciences & Center for Structural Biology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xinyu Zhao
- School of Pharmaceutical Sciences & Center for Structural Biology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lintao Song
- School of Pharmaceutical Sciences & Center for Structural Biology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Deli Zhuge
- School of Pharmaceutical Sciences & Center for Structural Biology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaokun Li
- School of Pharmaceutical Sciences & Center for Structural Biology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yingzheng Zhao
- School of Pharmaceutical Sciences & Center for Structural Biology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhifeng Huang
- School of Pharmaceutical Sciences & Center for Structural Biology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
23
|
Ebrahimikia Y, Darabi S, Rajaei F. Roles of stem cells in the treatment of Parkinson's disease. THE JOURNAL OF QAZVIN UNIVERSITY OF MEDICAL SCIENCES 2018. [DOI: 10.29252/qums.22.4.83] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
|
24
|
Jiménez KM, Pereira-Morales AJ, Adan A, Lopez-Leon S, Forero DA. Depressive symptoms are associated with a functional polymorphism in a miR-433 binding site in the FGF20 gene. Mol Brain 2018; 11:53. [PMID: 30241547 PMCID: PMC6151041 DOI: 10.1186/s13041-018-0397-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 09/18/2018] [Indexed: 01/30/2023] Open
Abstract
Genetic studies of major depressive disorder and its associated endophenotypes are useful for the identification of candidate genes. In recent years, variations in non-coding RNA genes, such as miRNAs, have been explored as novel candidates for psychiatric disorders and related endophenotypes. The aim of the present study was to evaluate the possible association between a functional polymorphism (rs12720208) in the FGF20 gene, which regulates its modulation by miR-433, and depressive symptoms in young adults. A sample of 270 participants from Colombia were evaluated with the Hospital Anxiety and Depression Scale - Depression Subscale (HADS-D) and genotyped for the rs12720208 polymorphism using a TaqMan assay. A lineal regression analysis was used. A statistically significant association of the functional polymorphism in the FGF20 gene (rs12720208) with depressive symptoms was found. It was observed that individuals with the G/A genotype had higher scores for the HADS-D subscale. Our results are the first description in the scientific literature about a significant association between a functional polymorphism in the FGF20 gene, which regulates its modulation by miR-433, and depressive symptoms.
Collapse
Affiliation(s)
- Karen M Jiménez
- Laboratory of Neuropsychiatric Genetics, Biomedical Sciences Research Group, School of Medicine, Universidad Antonio Nariño, 110231, Bogotá, Colombia
| | - Angela J Pereira-Morales
- Laboratory of Neuropsychiatric Genetics, Biomedical Sciences Research Group, School of Medicine, Universidad Antonio Nariño, 110231, Bogotá, Colombia
| | - Ana Adan
- Department of Clinical Psychology and Psychobiology, School of Psychology, University of Barcelona, Barcelona, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Sandra Lopez-Leon
- Novartis Pharmaceuticals Corporation, One Health Plaza, East Hanover, NJ, 07936-1080, USA.
| | - Diego A Forero
- Laboratory of Neuropsychiatric Genetics, Biomedical Sciences Research Group, School of Medicine, Universidad Antonio Nariño, 110231, Bogotá, Colombia.
| |
Collapse
|
25
|
Obergasteiger J, Frapporti G, Pramstaller PP, Hicks AA, Volta M. A new hypothesis for Parkinson's disease pathogenesis: GTPase-p38 MAPK signaling and autophagy as convergence points of etiology and genomics. Mol Neurodegener 2018; 13:40. [PMID: 30071902 PMCID: PMC6090926 DOI: 10.1186/s13024-018-0273-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 07/19/2018] [Indexed: 02/07/2023] Open
Abstract
The combination of genetics and genomics in Parkinson´s disease has recently begun to unveil molecular mechanisms possibly underlying disease onset and progression. In particular, catabolic processes such as autophagy have been increasingly gaining relevance as post-mortem evidence and experimental models suggested a participation in neurodegeneration and alpha-synuclein Lewy body pathology. In addition, familial Parkinson´s disease linked to LRRK2 and alpha-synuclein provided stronger correlation between etiology and alterations in autophagy. More detailed cellular pathways are proposed and genetic risk factors that associate with idiopathic Parkinson´s disease provide further clues in dissecting contributions of single players. Nevertheless, the fine-tuning of these processes remains elusive, as the initial stages of the pathways are not yet clarified.In this review, we collect literature evidence pointing to autophagy as the common, downstream target of Parkinsonian dysfunctions and augment current knowledge on the factors that direct the subsequent steps. Cell and molecular biology evidence indicate that p38 signaling underlies neurodegeneration and autoptic observations suggest a participation in neuropathology. Moreover, alpha-synuclein and LRRK2 also appear involved in the p38 pathway with additional roles in the regulation of GTPase signaling. Small GTPases are critical modulators of p38 activation and thus, their functional interaction with aSyn and LRRK2 could explain much of the detailed mechanics of autophagy in Parkinson´s disease.We propose a novel hypothesis for a more comprehensive working model where autophagy is controlled by upstream pathways, such as GTPase-p38, that have been so far underexplored in this context. In addition, etiological factors (LRRK2, alpha-synuclein) and risk loci might also combine in this common mechanism, providing a powerful experimental setting to dissect the cause of both familial and idiopathic disease.
Collapse
Affiliation(s)
- Julia Obergasteiger
- Institute for Biomedicine, Eurac Research – Affiliated Institute of the University of Lübeck, Via Galvani 31, 39100 Bolzano, Italy
| | - Giulia Frapporti
- Institute for Biomedicine, Eurac Research – Affiliated Institute of the University of Lübeck, Via Galvani 31, 39100 Bolzano, Italy
| | - Peter P. Pramstaller
- Institute for Biomedicine, Eurac Research – Affiliated Institute of the University of Lübeck, Via Galvani 31, 39100 Bolzano, Italy
- Department of Neurology, General Central Hospital, Via Böhler 5, 39100 Bolzano, Italy
- Department of Neurology, University of Lübeck, Ratzeburger Allee, 23538 Lübeck, Germany
| | - Andrew A. Hicks
- Institute for Biomedicine, Eurac Research – Affiliated Institute of the University of Lübeck, Via Galvani 31, 39100 Bolzano, Italy
| | - Mattia Volta
- Institute for Biomedicine, Eurac Research – Affiliated Institute of the University of Lübeck, Via Galvani 31, 39100 Bolzano, Italy
| |
Collapse
|
26
|
Shah SZA, Zhao D, Hussain T, Sabir N, Yang L. Regulation of MicroRNAs-Mediated Autophagic Flux: A New Regulatory Avenue for Neurodegenerative Diseases With Focus on Prion Diseases. Front Aging Neurosci 2018; 10:139. [PMID: 29867448 PMCID: PMC5962651 DOI: 10.3389/fnagi.2018.00139] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 04/25/2018] [Indexed: 02/06/2023] Open
Abstract
Prion diseases are fatal neurological disorders affecting various mammalian species including humans. Lack of proper diagnostic tools and non-availability of therapeutic remedies are hindering the control strategies for prion diseases. MicroRNAs (miRNAs) are abundant endogenous short non-coding essential RNA molecules that negatively regulate the target genes after transcription. Several biological processes depend on miRNAs, and altered profiles of these miRNAs are potential biomarkers for various neurodegenerative diseases, including prion diseases. Autophagic flux degrades the misfolded prion proteins to reduce chronic endoplasmic reticulum stress and enhance cell survival. Recent evidence suggests that specific miRNAs target and regulate the autophagic mechanism, which is critical for alleviating cellular stress. miRNAs-mediated regulation of these specific proteins involved in the autophagy represents a new target with highly significant therapeutic prospects. Here, we will briefly describe the biology of miRNAs, the use of miRNAs as potential biomarkers with their credibility, the regulatory mechanism of miRNAs in major neurodegenerative diseases such as Alzheimer’s, Parkinson’s, and prion diseases, degradation pathways for aggregated prion proteins, the role of autophagy in prion diseases. Finally, we will discuss the miRNAs-modulated autophagic flux in neurodegenerative diseases and employ them as potential therapeutic intervention strategy in prion diseases.
Collapse
Affiliation(s)
- Syed Zahid Ali Shah
- National Animal Transmissible Spongiform Encephalopathy Laboratory and Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Deming Zhao
- National Animal Transmissible Spongiform Encephalopathy Laboratory and Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Tariq Hussain
- National Animal Transmissible Spongiform Encephalopathy Laboratory and Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Naveed Sabir
- National Animal Transmissible Spongiform Encephalopathy Laboratory and Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Lifeng Yang
- National Animal Transmissible Spongiform Encephalopathy Laboratory and Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| |
Collapse
|
27
|
Single-Cell RNA-Seq of Mouse Dopaminergic Neurons Informs Candidate Gene Selection for Sporadic Parkinson Disease. Am J Hum Genet 2018; 102:427-446. [PMID: 29499164 DOI: 10.1016/j.ajhg.2018.02.001] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 02/01/2018] [Indexed: 12/22/2022] Open
Abstract
Genetic variation modulating risk of sporadic Parkinson disease (PD) has been primarily explored through genome-wide association studies (GWASs). However, like many other common genetic diseases, the impacted genes remain largely unknown. Here, we used single-cell RNA-seq to characterize dopaminergic (DA) neuron populations in the mouse brain at embryonic and early postnatal time points. These data facilitated unbiased identification of DA neuron subpopulations through their unique transcriptional profiles, including a postnatal neuroblast population and substantia nigra (SN) DA neurons. We use these population-specific data to develop a scoring system to prioritize candidate genes in all 49 GWAS intervals implicated in PD risk, including genes with known PD associations and many with extensive supporting literature. As proof of principle, we confirm that the nigrostriatal pathway is compromised in Cplx1-null mice. Ultimately, this systematic approach establishes biologically pertinent candidates and testable hypotheses for sporadic PD, informing a new era of PD genetic research.
Collapse
|
28
|
Zhou Y, Wang Z, Li J, Li X, Xiao J. Fibroblast growth factors in the management of spinal cord injury. J Cell Mol Med 2017; 22:25-37. [PMID: 29063730 PMCID: PMC5742738 DOI: 10.1111/jcmm.13353] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 07/12/2017] [Indexed: 12/11/2022] Open
Abstract
Spinal cord injury (SCI) possesses a significant health and economic burden worldwide. Traumatic SCI is a devastating condition that evolves through two successive stages. Throughout each of these stages, disturbances in ionic homeostasis, local oedema, ischaemia, focal haemorrhage, free radicals stress and inflammatory response were observed. Although there are no fully restorative cures available for SCI patients, various molecular, cellular and rehabilitative therapies, such as limiting local inflammation, preventing secondary cell death and enhancing the plasticity of local circuits in the spinal cord, were described. Current preclinical studies have showed that fibroblast growth factors (FGFs) alone or combination therapies utilizing cell transplantation and biomaterial scaffolds are proven effective for treating SCI in animal models. More importantly, some studies further demonstrated a paucity of clinical transfer usage to promote functional recovery of numerous patients with SCI. In this review, we focus on the therapeutic capacity and pitfalls of the FGF family and its clinical application for treating SCI, including the signalling component of the FGF pathway and the role in the central nervous system, the pathophysiology of SCI and the targets for FGF treatment. We also discuss the challenges and potential for the clinical translation of FGF-based approaches into treatments for SCI.
Collapse
Affiliation(s)
- Yulong Zhou
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.,Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhouguang Wang
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jiawei Li
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.,Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaokun Li
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jian Xiao
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.,Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
29
|
Tarale P, Daiwile AP, Sivanesan S, Stöger R, Bafana A, Naoghare PK, Parmar D, Chakrabarti T, Krishnamurthi K. Manganese exposure: Linking down-regulation of miRNA-7 and miRNA-433 with α-synuclein overexpression and risk of idiopathic Parkinson's disease. Toxicol In Vitro 2017; 46:94-101. [PMID: 28986288 DOI: 10.1016/j.tiv.2017.10.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 08/18/2017] [Accepted: 10/02/2017] [Indexed: 12/16/2022]
Abstract
Manganese is an essential trace element however elevated environmental and occupational exposure to this element has been correlated with neurotoxicity symptoms clinically identical to idiopathic Parkinson's disease. In the present study we chronically exposed human neuroblastoma SH-SY5Y cells to manganese (100μM) and carried out expression profiling of miRNAs known to modulate neuronal differentiation and neurodegeneration. The miRNA PCR array results reveal alterations in expression levels of miRNAs, which have previously been associated with the regulation of synaptic transmission and apoptosis. The expressions of miR-7 and miR-433 significantly reduced upon manganese exposure. By in silico homology analysis we identified SNCA and FGF-20as targets of miR-7 and miR-433. We demonstrate an inverse correlation in expression levels where reduction in these two miRNAs causes increases in SNCA and FGF-20. Transient transfection of SH-SY5Y cells with miR-7 and miR-433 mimics resulted in down regulation of SNCA and FGF-20 mRNA levels. Our study is the first to uncover the potential link between manganese exposure, altered miRNA expression and parkinsonism: manganese exposure causes overexpression of SNCA and FGF-20 by diminishing miR-7 and miR-433 levels. These miRNAs may be considered critical for protection from manganese induced neurotoxic mechanism and hence as potential therapeutic targets.
Collapse
Affiliation(s)
- Prashant Tarale
- Environmental Impact and Sustainability Division, CSIR-National Environmental Engineering Research Institute, Nagpur 440020, India; Schools of Biosciences, University of Nottingham, Sutton Bonington Campus, Leicestershire LE12 5RD, UK
| | - Atul P Daiwile
- Environmental Impact and Sustainability Division, CSIR-National Environmental Engineering Research Institute, Nagpur 440020, India
| | - Saravanadevi Sivanesan
- Environmental Impact and Sustainability Division, CSIR-National Environmental Engineering Research Institute, Nagpur 440020, India.
| | - Reinhard Stöger
- Schools of Biosciences, University of Nottingham, Sutton Bonington Campus, Leicestershire LE12 5RD, UK
| | - Amit Bafana
- Environmental Impact and Sustainability Division, CSIR-National Environmental Engineering Research Institute, Nagpur 440020, India
| | - Pravin K Naoghare
- Environmental Impact and Sustainability Division, CSIR-National Environmental Engineering Research Institute, Nagpur 440020, India
| | - Devendra Parmar
- Developmental Toxicology Division, CSIR-Indian Institute of Toxicology Research (IITR), Lucknow-226001, India
| | - Tapan Chakrabarti
- Visvesvaraya National Institute of Technology [VNIT], Nagpur 440010, India
| | - Kannan Krishnamurthi
- Environmental Impact and Sustainability Division, CSIR-National Environmental Engineering Research Institute, Nagpur 440020, India
| |
Collapse
|
30
|
Guo X, Liu T, Zhao D, Wang X, Liu D, He Y, Shan C, Kong Y, Hu W, Tao B, Sun L, Zhao H, Li S, Liu J. FGF18 protects against 6-hydroxydopamine-induced nigrostriatal damage in a rat model of Parkinson’s disease. Neuroscience 2017; 356:229-241. [DOI: 10.1016/j.neuroscience.2017.05.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 04/23/2017] [Accepted: 05/03/2017] [Indexed: 02/07/2023]
|
31
|
Genetic analysis of FGF20 in Chinese patients with Parkinson’s disease. Neurol Sci 2017; 38:887-891. [DOI: 10.1007/s10072-017-2868-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 02/21/2017] [Indexed: 11/27/2022]
|
32
|
Wang X, Sun X, Zhang X, Li H, Xie A. Quantitative assessment of the effect of FGF20 rs12720208 variant on the risk of Parkinson’s disease: a meta-analysis. Neurol Res 2017; 39:374-380. [PMID: 28191856 DOI: 10.1080/01616412.2017.1286542] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Xiaoli Wang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaoxuan Sun
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaona Zhang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hong Li
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Anmu Xie
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
33
|
Redenšek S, Trošt M, Dolžan V. Genetic Determinants of Parkinson's Disease: Can They Help to Stratify the Patients Based on the Underlying Molecular Defect? Front Aging Neurosci 2017; 9:20. [PMID: 28239348 PMCID: PMC5301007 DOI: 10.3389/fnagi.2017.00020] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 01/25/2017] [Indexed: 12/14/2022] Open
Abstract
Parkinson's disease (PD) is a sporadic progressive neurodegenerative brain disorder with a relatively strong genetic background. We have reviewed the current literature about the genetic factors that could be indicative of pathophysiological pathways of PD and their applications in everyday clinical practice. Information on novel risk genes is coming from several genome-wide association studies (GWASs) and their meta-analyses. GWASs that have been performed so far enabled the identification of 24 loci as PD risk factors. These loci take part in numerous cellular processes that may contribute to PD pathology: protein aggregation, protein, and membrane trafficking, lysosomal autophagy, immune response, synaptic function, endocytosis, inflammation, and metabolic pathways are among the most important ones. The identified single nucleotide polymorphisms are usually located in the non-coding regions and their functionality remains to be determined, although they presumably influence gene expression. It is important to be aware of a very low contribution of a single genetic risk factor to PD development; therefore, novel prognostic indices need to account for the cumulative nature of genetic risk factors. A better understanding of PD pathophysiology and its genetic background will help to elucidate the underlying pathological processes. Such knowledge may help physicians to recognize subjects with the highest risk for the development of PD, and provide an opportunity for the identification of novel potential targets for neuroprotective treatment. Moreover, it may enable stratification of the PD patients according to their genetic fingerprint to properly personalize their treatment as well as supportive measures.
Collapse
Affiliation(s)
- Sara Redenšek
- Pharmacogenetics Laboratory, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana Ljubljana, Slovenia
| | - Maja Trošt
- Department of Neurology, University Medical Centre Ljubljana Ljubljana, Slovenia
| | - Vita Dolžan
- Pharmacogenetics Laboratory, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana Ljubljana, Slovenia
| |
Collapse
|
34
|
Zhou WY, Zheng H, Du XL, Yang JL. Characterization of FGFR signaling pathway as therapeutic targets for sarcoma patients. Cancer Biol Med 2016; 13:260-8. [PMID: 27458533 PMCID: PMC4944539 DOI: 10.20892/j.issn.2095-3941.2015.0102] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The fibroblast growth factor receptor (FGFR) family plays important roles in regulating cell growth, proliferation, survival, differentiation and angiogenesis. Deregulation of the FGF/FGFR signaling pathway has been associated with multiple development syndromes and cancers, and thus therapeutic strategies targeting FGFs and FGFR in human cancer are currently being explored. However, few studies on the FGF/FGFR pathway have been conducted in sarcoma, which has a poor outcome with traditional treatments such as surgery, chemotherapy, and radiotherapy. Hence, in the present review, we provide an overview of the role of the FGF/FGFR pathway signal in sarcoma and FGFR inhibitors, which might be new targets for the treatment of sarcomas according to recent research.
Collapse
Affiliation(s)
| | - Hong Zheng
- Department of Epidemiology and Biostatistics, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
| | - Xiao-Ling Du
- Department of Diagnostics, Tianjin Medical University, Tianjin 300061, China
| | | |
Collapse
|
35
|
Cao DD, Li L, Chan WY. MicroRNAs: Key Regulators in the Central Nervous System and Their Implication in Neurological Diseases. Int J Mol Sci 2016; 17:E842. [PMID: 27240359 PMCID: PMC4926376 DOI: 10.3390/ijms17060842] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Revised: 05/20/2016] [Accepted: 05/23/2016] [Indexed: 01/03/2023] Open
Abstract
MicroRNAs (miRNAs) are a class of small, well-conserved noncoding RNAs that regulate gene expression post-transcriptionally. They have been demonstrated to regulate a lot of biological pathways and cellular functions. Many miRNAs are dynamically regulated during central nervous system (CNS) development and are spatially expressed in adult brain indicating their essential roles in neural development and function. In addition, accumulating evidence strongly suggests that dysfunction of miRNAs contributes to neurological diseases. These observations, together with their gene regulation property, implicated miRNAs to be the key regulators in the complex genetic network of the CNS. In this review, we first focus on the ways through which miRNAs exert the regulatory function and how miRNAs are regulated in the CNS. We then summarize recent findings that highlight the versatile roles of miRNAs in normal CNS physiology and their association with several types of neurological diseases. Subsequently we discuss the limitations of miRNAs research based on current studies as well as the potential therapeutic applications and challenges of miRNAs in neurological disorders. We endeavor to provide an updated description of the regulatory roles of miRNAs in normal CNS functions and pathogenesis of neurological diseases.
Collapse
Affiliation(s)
- Dan-Dan Cao
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong-Chinese Academy of Sciences Guangzhou Institute of Biomedicine and Health Joint Laboratory on Stem Cell and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong 999077, SAR, China.
| | - Lu Li
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong-Chinese Academy of Sciences Guangzhou Institute of Biomedicine and Health Joint Laboratory on Stem Cell and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong 999077, SAR, China.
| | - Wai-Yee Chan
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong-Chinese Academy of Sciences Guangzhou Institute of Biomedicine and Health Joint Laboratory on Stem Cell and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong 999077, SAR, China.
| |
Collapse
|
36
|
Turner CA, Eren-Koçak E, Inui EG, Watson SJ, Akil H. Dysregulated fibroblast growth factor (FGF) signaling in neurological and psychiatric disorders. Semin Cell Dev Biol 2016; 53:136-43. [PMID: 26454097 PMCID: PMC4833700 DOI: 10.1016/j.semcdb.2015.10.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 10/05/2015] [Indexed: 12/27/2022]
Abstract
The role of the fibroblast growth factor (FGF) system in brain-related disorders has received considerable attention in recent years. To understand the role of this system in neurological and psychiatric disorders, it is important to identify the specific members of the FGF family that are implicated, their location and the various mechanisms they can be modulated. Each disorder appears to impact specific molecular players in unique anatomical locations, and all of these could conceivably become targets for treatment. In the last several years, the issue of how to target this system directly has become an area of increasing interest. To date, the most promising therapeutics are small molecule inhibitors and antibodies that modulate FGF receptor (FGFR) function. Beyond attempting to modify the primary players affected by a given brain disorder, it may prove useful to target molecules, such as membrane-bound or extracellular proteins that interact with FGF ligands or FGFRs to modulate signaling.
Collapse
Affiliation(s)
- Cortney A Turner
- Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA.
| | - Emine Eren-Koçak
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey
| | | | - Stanley J Watson
- Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA; Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Huda Akil
- Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA; Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
37
|
Yuan L, Song Z, Deng X, Zheng W, Yang Z, Yang Y, Deng H. Genetic analysis of FGF20 variants in Chinese Han patients with essential tremor. Neurosci Lett 2016; 620:159-62. [PMID: 27040428 DOI: 10.1016/j.neulet.2016.03.055] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 03/15/2016] [Accepted: 03/31/2016] [Indexed: 12/22/2022]
Abstract
Essential tremor (ET) is one of the most frequent neurological disorders with elusive etiology, typically characterized by postural and kinetic tremors. Evidence reveals that genetic components are implicated in the development of ET and there are some overlaps between ET and Parkinson's disease in clinical features and etiology. Variants in the fibroblast growth factor 20 gene (FGF20) have been reported to be associated with the risk of Parkinson's disease. To evaluate the association between the FGF20 gene variants and ET susceptibility, we conducted genetic analysis of five FGF20 variants (rs1721100, rs1989754, rs10089600, rs12720208, and rs17550360) in 200 patients with ET and 426 ethnically-matched Chinese Han normal controls. Statistical analysis did not identify significant differences in genotypic or allelic frequencies of variants between ET patients and normal controls (all P>0.05). No related haplotype was found to be related to the risk of ET. The findings indicate the FGF20 gene might not play a dominating role in the genetic predisposition to ET in Chinese Han population.
Collapse
Affiliation(s)
- Lamei Yuan
- Center for Experimental Medicine, The Third Xiangya Hospital, Central South University, Changsha, China; Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhi Song
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiong Deng
- Center for Experimental Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Wen Zheng
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhijian Yang
- Center for Experimental Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yan Yang
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Hao Deng
- Center for Experimental Medicine, The Third Xiangya Hospital, Central South University, Changsha, China; Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
38
|
A WNT1-regulated developmental gene cascade prevents dopaminergic neurodegeneration in adult En1 mice. Neurobiol Dis 2015; 82:32-45. [DOI: 10.1016/j.nbd.2015.05.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 05/18/2015] [Accepted: 05/27/2015] [Indexed: 11/17/2022] Open
|
39
|
Gupta S, Verma S, Mantri S, Berman NE, Sandhir R. Targeting MicroRNAs in Prevention and Treatment of Neurodegenerative Disorders. Drug Dev Res 2015; 76:397-418. [PMID: 26359796 DOI: 10.1002/ddr.21277] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Preclinical Research microRNAs (miRNAs) are small noncoding RNAs (ncRNAs) that are key regulators of gene expression. They act on wide range of targets by binding to mRNA via imperfect complementarity at 3' UTR. Evidence suggests that miRNAs regulate many biological processes including neuronal development, differentiation, and disease. Altered expression of several miRNAs has been reported in many neurodegenerative disorders (NDDs). Many miRNAs are altered in these diseases, but miRNA 15, miRNA 21, and miRNA 146a have been shown to play critical role in many neurodegenerative conditions. As these miRNAs regulate many genes, miRNA targeted approaches would allow concurrently targeting of multiple effectors of pathways that regulate disease progression. In this review, we describe the role of miRNAs in various NDDs and their potential as therapeutic tools in prevention and treatment of neurological conditions.
Collapse
Affiliation(s)
- Smriti Gupta
- Department of Biochemistry, Panjab University, Chandigarh, 160014, India
| | - Savita Verma
- Department of Biochemistry, Panjab University, Chandigarh, 160014, India
| | - Shrikant Mantri
- Computational Biology Laboratory, National Agri-Food Biotechnology Institute, Mohali, Punjab, 160071, India
| | - Nancy E Berman
- Department of Anatomy & Cell Biology, Kansas University Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | - Rajat Sandhir
- Department of Biochemistry, Panjab University, Chandigarh, 160014, India
| |
Collapse
|
40
|
Ornitz DM, Itoh N. The Fibroblast Growth Factor signaling pathway. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2015; 4:215-66. [PMID: 25772309 PMCID: PMC4393358 DOI: 10.1002/wdev.176] [Citation(s) in RCA: 1435] [Impact Index Per Article: 143.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 11/23/2014] [Accepted: 01/08/2015] [Indexed: 12/13/2022]
Abstract
The signaling component of the mammalian Fibroblast Growth Factor (FGF) family is comprised of eighteen secreted proteins that interact with four signaling tyrosine kinase FGF receptors (FGFRs). Interaction of FGF ligands with their signaling receptors is regulated by protein or proteoglycan cofactors and by extracellular binding proteins. Activated FGFRs phosphorylate specific tyrosine residues that mediate interaction with cytosolic adaptor proteins and the RAS-MAPK, PI3K-AKT, PLCγ, and STAT intracellular signaling pathways. Four structurally related intracellular non-signaling FGFs interact with and regulate the family of voltage gated sodium channels. Members of the FGF family function in the earliest stages of embryonic development and during organogenesis to maintain progenitor cells and mediate their growth, differentiation, survival, and patterning. FGFs also have roles in adult tissues where they mediate metabolic functions, tissue repair, and regeneration, often by reactivating developmental signaling pathways. Consistent with the presence of FGFs in almost all tissues and organs, aberrant activity of the pathway is associated with developmental defects that disrupt organogenesis, impair the response to injury, and result in metabolic disorders, and cancer. For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- David M Ornitz
- Department of Developmental Biology, Washington University School of MedicineSt. Louis, MO, USA
- *
Correspondence to:
| | - Nobuyuki Itoh
- Graduate School of Pharmaceutical Sciences, Kyoto UniversitySakyo, Kyoto, Japan
| |
Collapse
|
41
|
Veenvliet JV, Smidt MP. Molecular mechanisms of dopaminergic subset specification: fundamental aspects and clinical perspectives. Cell Mol Life Sci 2014; 71:4703-27. [PMID: 25064061 PMCID: PMC11113784 DOI: 10.1007/s00018-014-1681-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 07/04/2014] [Accepted: 07/10/2014] [Indexed: 12/22/2022]
Abstract
Dopaminergic (DA) neurons in the ventral mesodiencephalon control locomotion and emotion and are affected in psychiatric and neurodegenerative diseases, such as Parkinson's disease (PD). A clinical hallmark of PD is the specific degeneration of DA neurons located within the substantia nigra (SNc), whereas neurons in the ventral tegmental area remain unaffected. Recent advances have highlighted that the selective vulnerability of the SNc may originate in subset-specific molecular programming during DA neuron development, and significantly increased our understanding of the molecular code that drives specific SNc development. We here present an up-to-date overview of molecular mechanisms that direct DA subset specification, integrating our current knowledge about subset-specific roles of transcription factors, signaling pathways and morphogenes. We discuss strategies to further unravel subset-specific gene-regulatory networks, and the clinical promise of fundamental knowledge about subset specification of DA neurons, with regards to cell replacement therapy and cell-type-specific vulnerability in PD.
Collapse
Affiliation(s)
- Jesse V. Veenvliet
- Department of Molecular Neuroscience, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| | - Marten P. Smidt
- Department of Molecular Neuroscience, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| |
Collapse
|
42
|
Tan L, Yu JT, Tan L. Causes and Consequences of MicroRNA Dysregulation in Neurodegenerative Diseases. Mol Neurobiol 2014; 51:1249-62. [PMID: 24973986 DOI: 10.1007/s12035-014-8803-9] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 06/22/2014] [Indexed: 12/12/2022]
Abstract
Neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD) and amyotrophic lateral sclerosis (ALS), originate from a loss of neurons in the central nervous system (CNS) and are severely debilitating. The incidence of neurodegenerative diseases increases with age, and they are expected to become more common due to extended life expectancy. Because of no clear mechanisms, these diseases have become a major challenge in neurobiology. It is well recognized that these disorders become the culmination of many different genetic and environmental influences. Prior studies have shown that microRNAs (miRNAs) are pathologically altered during the inexorable course of some neurodegenerative diseases, suggesting that miRNAs may be the contributing factor in neurodegeneration. Here, we review what is known about the involvement of miRNAs in the pathogenesis of neurodegenerative diseases. The biogenesis of miRNAs and various functions of miRNAs that act as the chief regulators will be discussed. We focus in particular on dysregulation of miRNAs which leads to several neurodegenerative diseases from three aspects: miRNA-generating disorders, miRNA-targeting genes and epigenetic alterations. Furthermore, recent evidences have shown that circulating miRNA expression levels are changed in patients with neurodegenerative diseases. Circulating miRNA expression levels are reported in patients in order to evaluate their application as biomarkers of these diseases. A discussion is included with a potential diagnostic biomarker and the possible future direction in exploring the nexus between miRNAs and various neurodegenerative diseases.
Collapse
Affiliation(s)
- Lin Tan
- College of Medicine and Pharmaceutics, Ocean University of China, Qingdao, China
| | | | | |
Collapse
|
43
|
Goodall EF, Heath PR, Bandmann O, Kirby J, Shaw PJ. Neuronal dark matter: the emerging role of microRNAs in neurodegeneration. Front Cell Neurosci 2013; 7:178. [PMID: 24133413 PMCID: PMC3794211 DOI: 10.3389/fncel.2013.00178] [Citation(s) in RCA: 139] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2013] [Accepted: 09/21/2013] [Indexed: 12/17/2022] Open
Abstract
MicroRNAs (miRNAs) are small, abundant RNA molecules that constitute part of the cell's non-coding RNA “dark matter.” In recent years, the discovery of miRNAs has revolutionised the traditional view of gene expression and our understanding of miRNA biogenesis and function has expanded. Altered expression of miRNAs is increasingly recognized as a feature of many disease states, including neurodegeneration. Here, we review the emerging role for miRNA dysfunction in Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis (ALS) and Huntington's disease pathogenesis. We emphasize the complex nature of gene regulatory networks and the need for systematic studies, with larger sample cohorts than have so far been reported, to reveal the most important miRNA regulators in disease. Finally, miRNA diversity and their potential to target multiple pathways, offers novel clinical applications for miRNAs as biomarkers and therapeutic agents in neurodegenerative diseases.
Collapse
Affiliation(s)
- Emily F Goodall
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience, University of Sheffield Sheffield, UK
| | | | | | | | | |
Collapse
|