1
|
Göksu AY. A review article on the development of dopaminergic neurons and establishment of dopaminergic neuron-based in vitro models by using immortal cell lines or stem cells to study and treat Parkinson's disease. Int J Dev Neurosci 2024; 84:817-842. [PMID: 39379284 DOI: 10.1002/jdn.10383] [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: 11/13/2023] [Revised: 09/09/2024] [Accepted: 09/12/2024] [Indexed: 10/10/2024] Open
Abstract
The primary pathological hallmark of Parkinson's disease (PD) is the degeneration of dopaminergic (DA) neurons in the substantia nigra pars compacta, a critical midbrain region. In vitro models based on DA neurons provide a powerful platform for investigating the cellular and molecular mechanisms of PD and testing novel therapeutic strategies. A deep understanding of DA neuron development, including the signalling pathways and transcription factors involved, is essential for advancing PD research. This article first explores the differentiation and maturation processes of DA neurons in the midbrain, detailing the relevant signalling pathways. It then compares various in vitro models, including primary cells, immortalized cell lines, and stem cell-based models, focusing on the advantages and limitations of each. Special attention is given to the role of immortalized and stem cell models in PD research. This review aims to guide researchers in selecting the most appropriate model for their specific research goals. Ethical considerations and clinical implications of using stem cells in PD research are also discussed.
Collapse
Affiliation(s)
- Azize Yasemin Göksu
- Department of Histology and Embryology, Department of Gene and Cell Therapy, Akdeniz University, School of Medicine, Antalya, Turkey
| |
Collapse
|
2
|
Liang C, Wei S, Ji Y, Lin J, Jiao W, Li Z, Yan F, Jing X. The role of enteric nervous system and GDNF in depression: Conversation between the brain and the gut. Neurosci Biobehav Rev 2024; 167:105931. [PMID: 39447778 DOI: 10.1016/j.neubiorev.2024.105931] [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: 06/26/2024] [Revised: 10/14/2024] [Accepted: 10/20/2024] [Indexed: 10/26/2024]
Abstract
Depression is a debilitating mental disorder that causes a persistent feeling of sadness and loss of interest. Approximately 280 million individuals worldwide suffer from depression by 2023. Despite the heavy medical and social burden imposed by depression, pathophysiology remains incompletely understood. Emerging evidence indicates various bidirectional interplay enable communication between the gut and brain. These interplays provide a link between intestinal and central nervous system as well as feedback from cortical and sensory centers to enteric activities, which also influences physiology and behavior in depression. This review aims to overview the significant role of the enteric nervous system (ENS) in the pathophysiology of depression and gut-brain axis's contribution to depressive disorders. Additionally, we explore the alterations in enteric glia cells (EGCs) and glial cell line-derived neurotrophic factor (GDNF) in depression and their involvement in neuronal support, intestinal homeostasis maintains and immune response activation. Modulating ENS function, EGCs and GDNF level could serve as novel strategies for future antidepressant therapy.
Collapse
Affiliation(s)
- Chuoyi Liang
- School of Nursing, Jinan University, Guangzhou, China
| | - Sijia Wei
- School of Nursing, Jinan University, Guangzhou, China
| | - Yelin Ji
- School of Nursing, Jinan University, Guangzhou, China
| | - Jiayi Lin
- School of Nursing, Jinan University, Guangzhou, China
| | - Wenli Jiao
- School of Nursing, Jinan University, Guangzhou, China
| | - Zhiying Li
- School of Nursing, Jinan University, Guangzhou, China
| | - Fengxia Yan
- School of Nursing, Jinan University, Guangzhou, China.
| | - Xi Jing
- School of Nursing, Jinan University, Guangzhou, China; Guangdong-Hong Kong-Macau Great Bay Area Geoscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, China.
| |
Collapse
|
3
|
Imraish A, Abu Thiab T, Alsalem M, Dahbour S, khleif H, Abu-Irmaileh B, Qasem R, El-Salem K. The neuroprotective effect of human primary astrocytes in multiple sclerosis: In vitro model. PLoS One 2024; 19:e0300203. [PMID: 38564643 PMCID: PMC10987000 DOI: 10.1371/journal.pone.0300203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 02/22/2024] [Indexed: 04/04/2024] Open
Abstract
Recent studies highlighted the role of astrocytes in neuroinflammatory diseases, particularly multiple sclerosis, interacting closely with other CNS components but also with the immune cells. However, due to the difficulty in obtaining human astrocytes, their role in these pathologies is still unclear. In this study we develop an astrocyte in vitro model to evaluate their role in multiple sclerosis after being treated with CSF isolated from both healthy and MS diagnosed patients. Gene expression and ELISA assays reveal that several pro-inflammatory markers IL-1β, TNF-α and IL-6, were significantly downregulated in astrocytes treated with MS-CSF. In contrast, neurotrophic survival, and growth factors, and GFAP, BDNF, GDNF and VEGF, were markedly elevated upon the same treatment. In summary, this study supports the notion of the astrocyte involvement in MS. The results reveal the neuroprotective role of astrocyte in MS pathogenicity by suppressing excessive inflammation and increasing the expression of tropic factors.
Collapse
Affiliation(s)
- Amer Imraish
- Department of Biological Sciences, School of Science, The University of Jordan, Amman, Jordan
| | - Tuqa Abu Thiab
- Department of Biological Sciences, School of Science, The University of Jordan, Amman, Jordan
| | - Mohammad Alsalem
- Department of Anatomy and Histology, School of Medicine, The University of Jordan, Amman, Jordan
| | - Saeed Dahbour
- Department of Neurology, Jordan University Hospital, The University of Jordan, Amman, Jordan
| | - Hiba khleif
- Department of Biological Sciences, School of Science, The University of Jordan, Amman, Jordan
| | | | - Raneen Qasem
- Department of Biological Sciences, School of Science, The University of Jordan, Amman, Jordan
| | - Khalid El-Salem
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
4
|
Kakoty V, Sarathlal KC, Kaur P, Wadhwa P, Vishwas S, Khan FR, Alhazmi AYM, Almasoudi HH, Gupta G, Chellappan DK, Paudel KR, Kumar D, Dua K, Singh SK. Unraveling the role of glial cell line-derived neurotrophic factor in the treatment of Parkinson's disease. Neurol Sci 2024; 45:1409-1418. [PMID: 38082050 DOI: 10.1007/s10072-023-07253-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 12/02/2023] [Indexed: 03/16/2024]
Abstract
Parkinson's disease is the second most common neurodegenerative condition with its prevalence projected to 8.9 million individuals globally in the year 2019. Parkinson's disease affects both motor and certain non-motor functions of an individual. Numerous research has focused on the neuroprotective effect of the glial cell line-derived neurotrophic factor (GDNF) in Parkinson's disease. Discovered in 1993, GDNF is a neurotrophic factor identified from the glial cells which was found to have selective effects on promoting survival and regeneration of certain populations of neurons including the dopaminergic nigrostriatal pathway. Given this property, recent studies have focused on the exogenous administration of GDNF for relieving Parkinson's disease-related symptoms both at a pre-clinical and a clinical level. This review will focus on enumerating the molecular connection between Parkinson's disease and GDNF and shed light on all the available drug delivery approaches to facilitate the selective delivery of GDNF into the brain paving the way as a potential therapeutic candidate for Parkinson's disease in the future.
Collapse
Affiliation(s)
- Violina Kakoty
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara, Punjab, India
| | - K C Sarathlal
- Department of Non-Communicable Disease, Translational Health Science and Technology Institute, Faridabad, India
| | - Palwinder Kaur
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara, Punjab, India
| | - Pankaj Wadhwa
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara, Punjab, India
| | - Sukriti Vishwas
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara, Punjab, India
| | - Farhan R Khan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Al-Quwayiyah, Shaqra University, Riyadh, Saudi Arabia
| | | | - Hassan Hussain Almasoudi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Najran University, Najran, 61441, Saudi Arabia
| | - Gaurav Gupta
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- School of Pharmacy, Graphic Era Hill University, Dehradun, 248007, India
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
| | | | - Keshav Raj Paudel
- Centre for Inflammation, Faculty of Science, School of Life Sciences, Centenary Institute and University of Technology Sydney, Sydney, NSW, 2050, Australia
| | - Dileep Kumar
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California Davis, One Shields Avenue, Davis, CA, 95616, USA
| | - Kamal Dua
- School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara, Punjab, India.
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
| |
Collapse
|
5
|
Kongsui R, Promsrisuk T, Klimaschewski L, Sriraksa N, Jittiwat J, Thongrong S. Pinostrobin mitigates neurodegeneration through an up-regulation of antioxidants and GDNF in a rat model of Parkinson's disease. F1000Res 2023; 12:846. [PMID: 38434672 PMCID: PMC10904945 DOI: 10.12688/f1000research.134891.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/23/2023] [Indexed: 03/05/2024] Open
Abstract
Background: One of the most common neurodegenerative diseases is Parkinson's disease (PD); PD is characterized by a reduction of neurons containing dopamine in the substantia nigra (SN), which leads to a lack of dopamine (DA) in nigrostriatal pathways, resulting in motor function disorders. Oxidative stress is considered as one of the etiologies involved in dopaminergic neuronal loss. Thus, we aimed to investigate the neuroprotective effects of pinostrobin (PB), a bioflavonoid extracted from Boesenbergia rotunda with antioxidative activity in PD. Methods: Rats were treated with 40 mg/kg of PB for seven consecutive days before and after 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD. After completing the experiment, the brains including SN and striatum were used for histological studies and biochemical assays. Results: PB treatment demonstrated a reduction of free radicals in the SN as indicated by significantly decreased MDA levels, whereas the antioxidative enzymes (SOD and GSH) were significantly increased. Furthermore, PB treatment significantly increased glial cell line-derived neurotrophic factor (GDNF) immunolabelling which has neurotrophic and neuroprotective effects on the survival of dopaminergic neurons. Furthermore, PB treatment was shown to protect CA1 and CA3 neurons in the hippocampus and dopaminergic neurons in the SN. DA levels in the SN were increased after PB treatment, leading to the improvement of motor function of PD rats. Conclusions: These results imply that PB prevents MPTP-induced neurotoxicity via its antioxidant activities and increases GDNF levels, which may contribute to the therapeutic strategy for PD.
Collapse
Affiliation(s)
- Ratchaniporn Kongsui
- Division of Physiology, School of Medical Sciences, University of Phayao, Mueang Phayao District, Phayao, 56000, Thailand
| | - Tichanon Promsrisuk
- Division of Physiology, School of Medical Sciences, University of Phayao, Mueang Phayao District, Phayao, 56000, Thailand
| | - Lars Klimaschewski
- Division of Neuroanatomy, Department of Anatomy Histology and Embryology, Innsbruck Medical University, Innsbruck, 6020, Austria
| | - Napatr Sriraksa
- Division of Physiology, School of Medical Sciences, University of Phayao, Mueang Phayao District, Phayao, 56000, Thailand
| | - Jinatta Jittiwat
- Faculty of Medicine, Mahasarakham University, Maha Sarakham, Maha Sarakham, 44000, Thailand
| | - Sitthisak Thongrong
- Division of Anatomy, School of Medical Sciences, University of Phayao, Mueang Phayao District, Phayao, 56000, Thailand
| |
Collapse
|
6
|
Li H, Dan QQ, Chen YJ, Chen L, Zhang HT, Mu DZ, Wang TH. Cellular Localization and Distribution of TGF-β1, GDNF and PDGF-BB in the Adult Primate Central Nervous System. Neurochem Res 2023; 48:2406-2423. [PMID: 36976393 DOI: 10.1007/s11064-023-03909-9] [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: 05/07/2022] [Revised: 03/03/2023] [Accepted: 03/08/2023] [Indexed: 03/29/2023]
Abstract
The available data on the localization of transforming growth factor beta1 (TGF-β1), glial cell line-derived neurotrophic factor (GDNF), and platelet-derived growth factor-BB (PDGF-BB) in the adult primate and human central nervous system (CNS) are limited and lack comprehensive and systematic information. This study aimed to investigate the cellular localization and distribution of TGF-β1, GDNF, and PDGF-BB in the CNS of adult rhesus macaque (Macaca mulatta). Seven adult rhesus macaques were included in the study. The protein levels of TGF-β1, PDGF-BB, and GDNF in the cerebral cortex, cerebellum, hippocampus, and spinal cord were analyzed by western blotting. The expression and location of TGF-β1, PDGF-BB, and GDNF in the brain and spinal cord was examined by immunohistochemistry and immunofluorescence staining, respectively. The mRNA expression of TGF-β1, PDGF-BB, and GDNF was detected by in situ hybridization. The molecular weight of TGF-β1, PDGF-BB, and GDNF in the homogenate of spinal cord was 25 KDa, 30 KDa, and 34 KDa, respectively. Immunolabeling revealed GDNF was ubiquitously distributed in the cerebral cortex, hippocampal formation, basal nuclei, thalamus, hypothalamus, brainstem, cerebellum, and spinal cord. TGF-β1 was least distributed and found only in the medulla oblongata and spinal cord, and PDGF-BB expression was also limited and present only in the brainstem and spinal cord. Besides, TGF-β1, PDGF-BB, and GDNF were localized in the astrocytes and microglia of spinal cord and hippocampus, and their expression was mainly found in the cytoplasm and primary dendrites. The mRNA of TGF-β1, PDGF-BB, and GDNF was localized to neuronal subpopulations in the spinal cord and cerebellum. These findings suggest that TGF-β1, GDNF and PDGF-BB may be associated with neuronal survival, neural regeneration and functional recovery in the CNS of adult rhesus macaques, providing the potential insights into the development or refinement of therapies based on these factors.
Collapse
Affiliation(s)
- Hui Li
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
- Department of Intensive Care Unit of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Qi-Qin Dan
- Institute of Neurological Disease, Sichuan University West China Hospital, Chengdu, Sichuan, 610041, China
| | - Yan-Jun Chen
- Institute of Neurological Disease, Sichuan University West China Hospital, Chengdu, Sichuan, 610041, China
| | - Li Chen
- Institute of Neurological Disease, Sichuan University West China Hospital, Chengdu, Sichuan, 610041, China
| | - Hong-Tian Zhang
- Institute of Neurological Disease, Sichuan University West China Hospital, Chengdu, Sichuan, 610041, China
| | - De-Zhi Mu
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.
| | - Ting-Hua Wang
- Institute of Neurological Disease, Sichuan University West China Hospital, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
7
|
Yeap YJ, Teddy TJW, Lee MJ, Goh M, Lim KL. From 2D to 3D: Development of Monolayer Dopaminergic Neuronal and Midbrain Organoid Cultures for Parkinson's Disease Modeling and Regenerative Therapy. Int J Mol Sci 2023; 24:ijms24032523. [PMID: 36768843 PMCID: PMC9917335 DOI: 10.3390/ijms24032523] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/24/2023] [Accepted: 01/26/2023] [Indexed: 01/31/2023] Open
Abstract
Parkinson's Disease (PD) is a prevalent neurodegenerative disorder that is characterized pathologically by the loss of A9-specific dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc) of the midbrain. Despite intensive research, the etiology of PD is currently unresolved, and the disease remains incurable. This, in part, is due to the lack of an experimental disease model that could faithfully recapitulate the features of human PD. However, the recent advent of induced pluripotent stem cell (iPSC) technology has allowed PD models to be created from patient-derived cells. Indeed, DA neurons from PD patients are now routinely established in many laboratories as monolayers as well as 3D organoid cultures that serve as useful toolboxes for understanding the mechanism underlying PD and also for drug discovery. At the same time, the iPSC technology also provides unprecedented opportunity for autologous cell-based therapy for the PD patient to be performed using the patient's own cells as starting materials. In this review, we provide an update on the molecular processes underpinning the development and differentiation of human pluripotent stem cells (PSCs) into midbrain DA neurons in both 2D and 3D cultures, as well as the latest advancements in using these cells for drug discovery and regenerative medicine. For the novice entering the field, the cornucopia of differentiation protocols reported for the generation of midbrain DA neurons may seem daunting. Here, we have distilled the essence of the different approaches and summarized the main factors driving DA neuronal differentiation, with the view to provide a useful guide to newcomers who are interested in developing iPSC-based models of PD.
Collapse
Affiliation(s)
- Yee Jie Yeap
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Tng J. W. Teddy
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
- Interdisciplinary Graduate Programme (IGP-Neuroscience), Nanyang Technological University, Singapore 639798, Singapore
| | - Mok Jung Lee
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Micaela Goh
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Kah Leong Lim
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
- National Neuroscience Institute, Singapore 308433, Singapore
- Department of Brain Sciences, Imperial College London, London SW7 2AZ, UK
- Department of Anatomy, Shanxi Medical University, Taiyuan 030001, China
- Correspondence:
| |
Collapse
|
8
|
Neuropathology of the Basal Ganglia in SNCA Transgenic Rat Model of Parkinson's Disease: Involvement of Parvalbuminergic Interneurons and Glial-Derived Neurotropic Factor. Int J Mol Sci 2022; 23:ijms231710126. [PMID: 36077524 PMCID: PMC9456397 DOI: 10.3390/ijms231710126] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 11/19/2022] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative disease characterized by the accumulation of alpha-synuclein, encoded by the SNCA gene. The main neuropathological hallmark of PD is the degeneration of dopaminergic neurons leading to striatal dopamine depletion. Trophic support by a neurotrophin called glial-derived neurotrophic factor (GDNF) is also lacking in PD. We performed immunohistochemical studies to investigate neuropathological changes in the basal ganglia of a rat transgenic model of PD overexpressing alfa-synuclein. We observed that neuronal loss also occurs in the dorsolateral part of the striatum in the advanced stages of the disease. Moreover, along with the degeneration of the medium spiny projection neurons, we found a dramatic loss of parvalbumin interneurons. A marked decrease in GDNF, which is produced by parvalbumin interneurons, was observed in the striatum and in the substantia nigra of these animals. This confirmed the involvement of the striatum in the pathophysiology of PD and the importance of GDNF in maintaining the health of the substantia nigra.
Collapse
|
9
|
Razenkova VA, Korzhevskii DE. Catecholaminergic Rat’s Forebrain Structures in Early Postnatal Development and Aging. Russ J Dev Biol 2022. [DOI: 10.1134/s1062360422030067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
10
|
Reawakening GDNF's regenerative past in mice and humans. Regen Ther 2022; 20:78-85. [PMID: 35509264 PMCID: PMC9043678 DOI: 10.1016/j.reth.2022.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/18/2022] [Accepted: 03/31/2022] [Indexed: 11/22/2022] Open
Abstract
The ability of an animal to regenerate lost tissue and body parts has obviously life-saving implications. Understanding how this ability became restricted or active in specific animal lineages will help us understand our own regeneration. According to phylogenic analysis, the glial cell line-derived neurotrophic factor (GDNF) signaling pathway, but not other family members, is conserved in axolotls, a salamander with remarkable regenerative capacity. Furthermore, comparing the pro-regenerative Spiny mouse to its less regenerative descendant, the House mouse, revealed that the GDNF signaling pathway, but not other family members, was induced in regenerating Spiny mice. According to GDNF receptor expression analysis, GDNF may promote hair follicle neogenesis – an important feature of skin regeneration – by determining the fate of dermal fibroblasts as part of new hair follicles. These findings support the idea that GDNF treatment will promote skin regeneration in humans by demonstrating the GDNF signaling pathway's ancestral and cellular nature. In pro-regenerative axolotls, the GDNF-GFR□1 signaling system is conserved. In pro-regenerative Spiny mice, the GDNF-GFR□1 signaling system is activated. In mice, GDNF targets upper-regeneration-competent dermal fibroblasts. GDNF-GFR□1 activation may promote skin regeneration in mice and humans.
Collapse
|
11
|
Lu J, Lu L, Yu Y, Oliphant K, Drobyshevsky A, Claud EC. Early preterm infant microbiome impacts adult learning. Sci Rep 2022; 12:3310. [PMID: 35228616 PMCID: PMC8885646 DOI: 10.1038/s41598-022-07245-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 02/10/2022] [Indexed: 12/14/2022] Open
Abstract
Interventions to mitigate long-term neurodevelopmental deficits such as memory and learning impairment in preterm infants are warranted. Manipulation of the gut microbiome affects host behaviors. In this study we determined whether early maturation of the infant microbiome is associated with neurodevelopment outcomes. Germ free mice colonized at birth with human preterm infant microbiomes from infants of advancing post menstrual age (PMA) demonstrated an increase in bacterial diversity and a shift in dominance of taxa mimicking the human preterm microbiome development pattern. These characteristics along with changes in a number of metabolites as the microbiome matured influenced associative learning and memory but not locomotor ability, anxiety-like behaviors, or social interaction in adult mice. As a regulator of learning and memory, brain glial cell-derived neurotrophic factor increased with advancing PMA and was also associated with better performance in associative learning and memory in adult mice. We conclude that maturation of the microbiome in early life of preterm infants primes adult associative memory and learning ability. Our findings suggest a critical window of early intervention to affect maturation of the preterm infant microbiome and ultimately improve neurodevelopmental outcomes.
Collapse
Affiliation(s)
- Jing Lu
- Department of Pediatrics, Pritzker School of Medicine/Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Lei Lu
- Department of Pediatrics, Pritzker School of Medicine/Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Yueyue Yu
- Department of Pediatrics, Pritzker School of Medicine/Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Kaitlyn Oliphant
- Department of Pediatrics, Pritzker School of Medicine/Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Alexander Drobyshevsky
- Department of Pediatrics, NorthShore University HealthSystem Research Institute, Evanston, IL, 60202, USA
| | - Erika C Claud
- Department of Pediatrics, Pritzker School of Medicine/Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
12
|
Huang M, Xu L, Liu J, Huang P, Tan Y, Chen S. Cell–Cell Communication Alterations via Intercellular Signaling Pathways in Substantia Nigra of Parkinson’s Disease. Front Aging Neurosci 2022; 14:828457. [PMID: 35283752 PMCID: PMC8914319 DOI: 10.3389/fnagi.2022.828457] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 01/17/2022] [Indexed: 11/13/2022] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative movement disorder characterized with dopaminergic neuron (DaN) loss within the substantia nigra (SN). Despite bulk studies focusing on intracellular mechanisms of PD inside DaNs, few studies have explored the pathogeneses outside DaNs, or between DaNs and other cells. Here, we set out to probe the implication of intercellular communication involving DaNs in the pathogeneses of PD at a systemic level with bioinformatics methods. We harvested three online published single-cell/single-nucleus transcriptomic sequencing (sc/snRNA-seq) datasets of human SN (GSE126838, GSE140231, and GSE157783) from the Gene Expression Omnibus (GEO) database, and integrated them with one of the latest integration algorithms called Harmony. We then applied CellChat, the latest cell–cell communication analytic algorithm, to our integrated dataset. We first found that the overall communication quantity was decreased while the overall communication strength was enhanced in PD sample compared with control sample. We then focused on the intercellular communication where DaNs are involved, and found that the communications between DaNs and other cell types via certain signaling pathways were selectively altered in PD, including some growth factors, neurotrophic factors, chemokines, etc. pathways. Our bioinformatics analysis showed that the alteration in intercellular communications involving DaNs might be a previously underestimated aspect of PD pathogeneses with novel translational potential.
Collapse
Affiliation(s)
- Maoxin Huang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liang Xu
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jin Liu
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pei Huang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuyan Tan
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Yuyan Tan,
| | - Shengdi Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Lab for Translational Research of Neurodegenerative Diseases, Shanghai Institute for Advanced Immunochemical Studies, Shanghai Tech University, Shanghai, China
- Shengdi Chen,
| |
Collapse
|
13
|
Revishchin AV, Parshina VV, Pavlova GV. [The role of glial cell line-derived neurotrophic factor isoforms in human glial tumors]. ZHURNAL VOPROSY NEIROKHIRURGII IMENI N. N. BURDENKO 2022; 86:106-112. [PMID: 36534631 DOI: 10.17116/neiro202286061106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Glial cell line-derived neurotrophic factor (GDNF) has a wide range of actions and positively affects viability, proliferative activity and migratory ability of cells in nervous system. That is why GDNF is being considered as a therapeutic molecule in the treatment of neurodegenerative diseases, in particular Parkinson's disease. However, GDNF has the same effect on high-grade glioma cells promoting their growth, resistance to therapy and dissemination. Expression of this factor in tissues and cultures of gliomas is up to five times higher than in intact brain matter. It was revealed that epigenetic modifications in GDNF gene promoter contribute to overexpression. Target suppression of GDNF gene transcription slows down growth of glioma and decreases cell migration. This review is devoted to the effect of GDNF on glioma cells, causes and consequences of its overexpression. Further analysis of expression and function of various GDNF isoforms in glial tumors may be valuable to develop new treatment methods for these dangerous diseases.
Collapse
Affiliation(s)
- A V Revishchin
- Institute of Higher Nervous Activity and Neurophysiology, Moscow, Russia
| | - V V Parshina
- Institute of Higher Nervous Activity and Neurophysiology, Moscow, Russia
- Burdenko Neurosurgical Center, Moscow, Russia
| | - G V Pavlova
- Institute of Higher Nervous Activity and Neurophysiology, Moscow, Russia
- Burdenko Neurosurgical Center, Moscow, Russia
- Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
14
|
Sakakibara Y, Hirota Y, Ibaraki K, Takei K, Chikamatsu S, Tsubokawa Y, Saito T, Saido TC, Sekiya M, Iijima KM. Widespread Reduced Density of Noradrenergic Locus Coeruleus Axons in the App Knock-In Mouse Model of Amyloid-β Amyloidosis. J Alzheimers Dis 2021; 82:1513-1530. [PMID: 34180416 PMCID: PMC8461671 DOI: 10.3233/jad-210385] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND The locus coeruleus (LC), a brainstem nucleus comprising noradrenergic neurons, is one of the earliest regions affected by Alzheimer's disease (AD). Amyloid-β (Aβ) pathology in the cortex in AD is thought to exacerbate the age-related loss of LC neurons, which may lead to cortical tau pathology. However, mechanisms underlying LC neurodegeneration remain elusive. OBJECTIVE Here, we aimed to examine how noradrenergic neurons are affected by cortical Aβ pathology in AppNL-G-F/NL-G-F knock-in mice. METHODS The density of noradrenergic axons in LC-innervated regions and the LC neuron number were analyzed by an immunohistochemical method. To explore the potential mechanisms for LC degeneration, we also examined the occurrence of tau pathology in LC neurons, the association of reactive gliosis with LC neurons, and impaired trophic support in the brains of AppNL-G-F/NL-G-F mice. RESULTS We observed a significant reduction in the density of noradrenergic axons from the LC in aged AppNL-G-F/NL-G-F mice without neuron loss or tau pathology, which was not limited to areas near Aβ plaques. However, none of the factors known to be related to the maintenance of LC neurons (i.e., somatostatin/somatostatin receptor 2, brain-derived neurotrophic factor, nerve growth factor, and neurotrophin-3) were significantly reduced in AppNL-G-F/NL-G-F mice. CONCLUSION This study demonstrates that cortical Aβ pathology induces noradrenergic neurodegeneration, and further elucidation of the underlying mechanisms will reveal effective therapeutics to halt AD progression.
Collapse
Affiliation(s)
- Yasufumi Sakakibara
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Yu Hirota
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Kyoko Ibaraki
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Kimi Takei
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Sachie Chikamatsu
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Yoko Tsubokawa
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Saitama, Japan
| | - Michiko Sekiya
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan.,Department of Experimental Gerontology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Koichi M Iijima
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan.,Department of Experimental Gerontology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| |
Collapse
|
15
|
Li X, Li K, Chen Y, Fang F. The Role of Hippo Signaling Pathway in the Development of the Nervous System. Dev Neurosci 2021; 43:263-270. [PMID: 34350875 DOI: 10.1159/000515633] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 02/26/2021] [Indexed: 11/19/2022] Open
Abstract
Hippo signaling pathway is a highly conserved and crucial signaling pathway that controls the size of tissues and organs by regulating the proliferation, differentiation, and apoptosis of cells. The nervous system is a complicated system that participates in information collection, integration, and procession. The balance of various aspects of the nervous system is vital for the normal regulation of physiological conditions of the body, like the population and distribution of nerve cells, nerve connections, and so on. Defects in these aspects may lead to cognitive, behavioral, and neurological dysfunction, resulting in various nervous system diseases. Recently, accumulating evidence proposes that Hippo pathway maintains numerous biological functions in the nervous system development, including modulating the proliferation and differentiation of nerve cells and promoting the development of synapse, corpus callosum, and cortex. In this review, we will summarize recent findings of Hippo pathway in the nervous system to improve our understanding on its function and to provide potential therapeutic strategies of nervous system diseases in the future.
Collapse
Affiliation(s)
- Xifan Li
- Department of Human Anatomy, School of Basic Medicine Sciences, Guilin Medical University, Guilin, China
| | - Kaixuan Li
- Department of Human Anatomy, School of Basic Medicine Sciences, Guilin Medical University, Guilin, China
| | - Yu Chen
- Department of Human Anatomy, School of Basic Medicine Sciences, Guilin Medical University, Guilin, China
| | - Fang Fang
- Department of Human Anatomy, School of Basic Medicine Sciences, Guilin Medical University, Guilin, China
| |
Collapse
|
16
|
Troncoso-Escudero P, Sepulveda D, Pérez-Arancibia R, Parra AV, Arcos J, Grunenwald F, Vidal RL. On the Right Track to Treat Movement Disorders: Promising Therapeutic Approaches for Parkinson's and Huntington's Disease. Front Aging Neurosci 2020; 12:571185. [PMID: 33101007 PMCID: PMC7497570 DOI: 10.3389/fnagi.2020.571185] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/17/2020] [Indexed: 12/17/2022] Open
Abstract
Movement disorders are neurological conditions in which patients manifest a diverse range of movement impairments. Distinct structures within the basal ganglia of the brain, an area involved in movement regulation, are differentially affected for every disease. Among the most studied movement disorder conditions are Parkinson's (PD) and Huntington's disease (HD), in which the deregulation of the movement circuitry due to the loss of specific neuronal populations in basal ganglia is the underlying cause of motor symptoms. These symptoms are due to the loss principally of dopaminergic neurons of the substantia nigra (SN) par compacta and the GABAergic neurons of the striatum in PD and HD, respectively. Although these diseases were described in the 19th century, no effective treatment can slow down, reverse, or stop disease progression. Available pharmacological therapies have been focused on preventing or alleviating motor symptoms to improve the quality of life of patients, but these drugs are not able to mitigate the progressive neurodegeneration. Currently, considerable therapeutic advances have been achieved seeking a more efficacious and durable therapeutic effect. Here, we will focus on the new advances of several therapeutic approaches for PD and HD, starting with the available pharmacological treatments to alleviate the motor symptoms in both diseases. Then, we describe therapeutic strategies that aim to restore specific neuronal populations or their activity. Among the discussed strategies, the use of Neurotrophic factors (NTFs) and genetic approaches to prevent the neuronal loss in these diseases will be described. We will highlight strategies that have been evaluated in both Parkinson's and Huntington's patients, and also the ones with strong preclinical evidence. These current therapeutic techniques represent the most promising tools for the safe treatment of both diseases, specifically those aimed to avoid neuronal loss during disease progression.
Collapse
Affiliation(s)
- Paulina Troncoso-Escudero
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Faculty of Medicine, Biomedical Neuroscience Institute, University of Chile, Santiago, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health, and Metabolism, University of Chile, Santiago, Chile
| | - Denisse Sepulveda
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Faculty of Medicine, Biomedical Neuroscience Institute, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health, and Metabolism, University of Chile, Santiago, Chile
| | - Rodrigo Pérez-Arancibia
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Faculty of Medicine, Biomedical Neuroscience Institute, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health, and Metabolism, University of Chile, Santiago, Chile
| | - Alejandra V. Parra
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Faculty of Medicine, Biomedical Neuroscience Institute, University of Chile, Santiago, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health, and Metabolism, University of Chile, Santiago, Chile
| | - Javiera Arcos
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Faculty of Medicine, Biomedical Neuroscience Institute, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health, and Metabolism, University of Chile, Santiago, Chile
| | - Felipe Grunenwald
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Faculty of Medicine, Biomedical Neuroscience Institute, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health, and Metabolism, University of Chile, Santiago, Chile
| | - Rene L. Vidal
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Faculty of Medicine, Biomedical Neuroscience Institute, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health, and Metabolism, University of Chile, Santiago, Chile
| |
Collapse
|
17
|
Enterría-Morales D, Del Rey NLG, Blesa J, López-López I, Gallet S, Prévot V, López-Barneo J, d'Anglemont de Tassigny X. Molecular targets for endogenous glial cell line-derived neurotrophic factor modulation in striatal parvalbumin interneurons. Brain Commun 2020; 2:fcaa105. [PMID: 32954345 PMCID: PMC7472905 DOI: 10.1093/braincomms/fcaa105] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 06/05/2020] [Accepted: 06/23/2020] [Indexed: 12/20/2022] Open
Abstract
Administration of recombinant glial cell line-derived neurotrophic factor into the putamen has been tested in preclinical and clinical studies to evaluate its neuroprotective effects on the progressive dopaminergic neuronal degeneration that characterizes Parkinson’s disease. However, intracerebral glial cell line-derived neurotrophic factor infusion is a challenging therapeutic strategy, with numerous potential technical and medical limitations. Most of these limitations could be avoided if the production of endogenous glial cell line-derived neurotrophic factor could be increased. Glial cell line-derived neurotrophic factor is naturally produced in the striatum from where it exerts a trophic action on the nigrostriatal dopaminergic pathway. Most of striatal glial cell line-derived neurotrophic factor is synthesized by a subset of GABAergic interneurons characterized by the expression of parvalbumin. We sought to identify molecular targets specific to those neurons and which are putatively associated with glial cell line-derived neurotrophic factor synthesis. To this end, the transcriptomic differences between glial cell line-derived neurotrophic factor-positive parvalbumin neurons in the striatum and parvalbumin neurons located in the nearby cortex, which do not express glial cell line-derived neurotrophic factor, were analysed. Using mouse reporter models, we have defined the genomic signature of striatal parvalbumin interneurons obtained by fluorescence-activated cell sorting followed by microarray comparison. Short-listed genes were validated by additional histological and molecular analyses. These genes code for membrane receptors (Kit, Gpr83, Tacr1, Tacr3, Mc3r), cytosolic proteins (Pde3a, Crabp1, Rarres2, Moxd1) and a transcription factor (Lhx8). We also found the proto-oncogene cKit to be highly specific of parvalbumin interneurons in the non-human primate striatum, thus highlighting a conserved expression between species and suggesting that specific genes identified in mouse parvalbumin neurons could be putative targets in the human brain. Pharmacological stimulation of four G-protein-coupled receptors enriched in the striatal parvalbumin interneurons inhibited Gdnf expression presumably by decreasing cyclic adenosine monophosphate formation. Additional experiments with pharmacological modulators of adenylyl cyclase and protein kinase A indicated that this pathway is a relevant intracellular route to induce Gdnf gene activation. This preclinical study is an important step in the ongoing development of a specific pro-endo-glial cell line-derived neurotrophic factor pharmacological strategy to treat Parkinson’s disease.
Collapse
Affiliation(s)
- Daniel Enterría-Morales
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain.,Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
| | | | - Javier Blesa
- HM CINAC, Hospital Universitario HM Puerta del Sur, Móstoles, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Ivette López-López
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain
| | - Sarah Gallet
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, Laboratory of Development and Plasticity of the Neuroendocrine Brain, UMR-S 1172, Lille, France
| | - Vincent Prévot
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, Laboratory of Development and Plasticity of the Neuroendocrine Brain, UMR-S 1172, Lille, France
| | - José López-Barneo
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain.,Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Xavier d'Anglemont de Tassigny
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain.,Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
18
|
Molecular Regulation in Dopaminergic Neuron Development. Cues to Unveil Molecular Pathogenesis and Pharmacological Targets of Neurodegeneration. Int J Mol Sci 2020; 21:ijms21113995. [PMID: 32503161 PMCID: PMC7312927 DOI: 10.3390/ijms21113995] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/30/2020] [Accepted: 06/01/2020] [Indexed: 12/13/2022] Open
Abstract
The relatively few dopaminergic neurons in the mammalian brain are mostly located in the midbrain and regulate many important neural functions, including motor integration, cognition, emotive behaviors and reward. Therefore, alteration of their function or degeneration leads to severe neurological and neuropsychiatric diseases. Unraveling the mechanisms of midbrain dopaminergic (mDA) phenotype induction and maturation and elucidating the role of the gene network involved in the development and maintenance of these neurons is of pivotal importance to rescue or substitute these cells in order to restore dopaminergic functions. Recently, in addition to morphogens and transcription factors, microRNAs have been identified as critical players to confer mDA identity. The elucidation of the gene network involved in mDA neuron development and function will be crucial to identify early changes of mDA neurons that occur in pre-symptomatic pathological conditions, such as Parkinson’s disease. In addition, it can help to identify targets for new therapies and for cell reprogramming into mDA neurons. In this essay, we review the cascade of transcriptional and posttranscriptional regulation that confers mDA identity and regulates their functions. Additionally, we highlight certain mechanisms that offer important clues to unveil molecular pathogenesis of mDA neuron dysfunction and potential pharmacological targets for the treatment of mDA neuron dysfunction.
Collapse
|
19
|
Padmakumar S, Taha MS, Kadakia E, Bleier BS, Amiji MM. Delivery of neurotrophic factors in the treatment of age-related chronic neurodegenerative diseases. Expert Opin Drug Deliv 2020; 17:323-340. [DOI: 10.1080/17425247.2020.1727443] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Smrithi Padmakumar
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA, USA
| | - Maie S. Taha
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA, USA
- Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Ekta Kadakia
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA, USA
- Drug Metabolism and Pharmacokinetics (DMPK), Biogen Inc, Cambridge, MA, USA
| | - Benjamin S. Bleier
- Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
| | - Mansoor M. Amiji
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA, USA
| |
Collapse
|
20
|
Duarte Azevedo M, Sander S, Tenenbaum L. GDNF, A Neuron-Derived Factor Upregulated in Glial Cells during Disease. J Clin Med 2020; 9:E456. [PMID: 32046031 PMCID: PMC7073520 DOI: 10.3390/jcm9020456] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/31/2020] [Accepted: 02/03/2020] [Indexed: 12/20/2022] Open
Abstract
In a healthy adult brain, glial cell line-derived neurotrophic factor (GDNF) is exclusively expressed by neurons, and, in some instances, it has also been shown to derive from a single neuronal subpopulation. Secreted GDNF acts in a paracrine fashion by forming a complex with the GDNF family receptor α1 (GFRα1), which is mainly expressed by neurons and can act in cis as a membrane-bound factor or in trans as a soluble factor. The GDNF/GFRα1 complex signals through interactions with the "rearranged during transfection" (RET) receptor or via the neural cell adhesion molecule (NCAM) with a lower affinity. GDNF can also signal independently from GFRα1 by interacting with syndecan-3. RET, which is expressed by neurons involved in several pathways (nigro-striatal dopaminergic neurons, motor neurons, enteric neurons, sensory neurons, etc.), could be the main determinant of the specificity of GDNF's pro-survival effect. In an injured brain, de novo expression of GDNF occurs in glial cells. Neuroinflammation has been reported to induce GDNF expression in activated astrocytes and microglia, infiltrating macrophages, nestin-positive reactive astrocytes, and neuron/glia (NG2) positive microglia-like cells. This disease-related GDNF overexpression can be either beneficial or detrimental depending on the localization in the brain and the level and duration of glial cell activation. Some reports also describe the upregulation of RET and GFRα1 in glial cells, suggesting that GDNF could modulate neuroinflammation.
Collapse
Affiliation(s)
| | | | - Liliane Tenenbaum
- Laboratory of Molecular Neurotherapies and NeuroModulation, Center for Neuroscience Research, Lausanne University Hospital, CHUV-Pavillon 3, av de Beaumont, CH-1010 Lausanne, Switzerland; (M.D.A.); (S.S.)
| |
Collapse
|
21
|
Enterría-Morales D, López-López I, López-Barneo J, d'Anglemont de Tassigny X. Role of Glial Cell Line-Derived Neurotrophic Factor in the Maintenance of Adult Mesencephalic Catecholaminergic Neurons. Mov Disord 2020; 35:565-576. [PMID: 31930748 DOI: 10.1002/mds.27986] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 12/16/2019] [Accepted: 12/27/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The glial cell line-derived neurotrophic factor has a potent neuroprotective action on mesencephalic dopamine neurons, which are progressively lost in Parkinson's disease. Intrastriatal administration of this factor is a promising therapy for Parkinson's disease. Glial cell line-derived neurotrophic factor is naturally produced in restricted cerebral regions, such as the striatum, septum, and thalamus; however, its effects in the adult brain remain under debate. OBJECTIVES We sought to clarify the physiologic role of endogenous glial cell line-derived neurotrophic factor in the survival of catecholaminergic neurons of the substantia nigra pars compacta and the locus coeruleus in adult mice. METHODS We used 2 new Cre recombinase-based mouse models to delete a floxed-glial cell line-derived neurotrophic factor gene. The first model had Cre expression in the parvalbumin expressing interneurons, as these cells represent the major source of striatal glial cell line-derived neurotrophic factor. The second model was an estrogen receptor 2-based inducible Cre triggered by tamoxifen at 2 months of age. RESULTS We found that the floxed-glial cell line-derived neurotrophic factor gene was resilient to ablation by Cre-induced recombination and that parvalbumin-driven Cre was particularly inefficient to do so. The inducible-Cre model allowed an average 70% to 80% reduction in glial cell line-derived neurotrophic factor messenger ribonucleic acid and protein in striatum and septum with moderate significant loss of catecholamine neurons in the nigrostriatal pathway and, more markedly, in the locus coeruleus. This was accompanied with mild locomotor decline. CONCLUSIONS Our data support qualitatively the view that brain glial cell line-derived neurotrophic factor is needed for the maintenance of adult central catecholaminergic neurons. © 2020 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Daniel Enterría-Morales
- Institute of Biomedicine of Seville, Virgen del Rocío University Hospital,/CSIC/University of Seville, Seville, Spain.,Network Center for Biomedical Research on Neurodegenerative Diseases, Seville, Spain
| | - Ivette López-López
- Institute of Biomedicine of Seville, Virgen del Rocío University Hospital,/CSIC/University of Seville, Seville, Spain.,Network Center for Biomedical Research on Neurodegenerative Diseases, Seville, Spain
| | - José López-Barneo
- Institute of Biomedicine of Seville, Virgen del Rocío University Hospital,/CSIC/University of Seville, Seville, Spain.,Network Center for Biomedical Research on Neurodegenerative Diseases, Seville, Spain
| | - Xavier d'Anglemont de Tassigny
- Institute of Biomedicine of Seville, Virgen del Rocío University Hospital,/CSIC/University of Seville, Seville, Spain.,Network Center for Biomedical Research on Neurodegenerative Diseases, Seville, Spain
| |
Collapse
|
22
|
Furukawa Y, Hara RI, Nakaya M, Okuyama S, Sawamoto A, Nakajima M. Citrus Auraptene Induces Glial Cell Line-Derived Neurotrophic Factor in C6 Cells. Int J Mol Sci 2019; 21:ijms21010253. [PMID: 31905925 PMCID: PMC6981972 DOI: 10.3390/ijms21010253] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/24/2019] [Accepted: 12/27/2019] [Indexed: 12/12/2022] Open
Abstract
We previously demonstrated that auraptene (AUR), a natural coumarin derived from citrus plants, exerts anti-inflammatory effects in the brain, resulting in neuroprotection in some mouse models of brain disorders. The present study showed that treatment with AUR significantly increased the release of glial cell line-derived neurotrophic factor (GDNF), in a dose- and time-dependent manner, by rat C6 glioma cells, which release was associated with increased expression of GDNF mRNA. These results suggest that AUR acted as a neuroprotective agent in the brain via not only its anti-inflammatory action but also its induction of neurotrophic factor. We also showed that (1) the AUR-induced GDNF production was inhibited by U0126, a specific inhibitor of mitogen-activated protein kinase/extracellular signal-regulated kinase (ERK) 1/2, and by H89, a specific inhibitor of protein kinase A (PKA); and (2) AUR induced the phosphorylation of cAMP response element-binding protein (CREB), a transcription factor located within the nucleus. These results suggest that AUR-stimulated gdnf gene expression was up-regulated through the PKA/ERK/CREB pathway in C6 cells.
Collapse
|
23
|
Wang S, Fan Y, Xu Y, Zhang L, Cai L, Lv B. GDNFOS1 knockdown decreases the invasion and viability of glioblastoma cells. Exp Ther Med 2019; 18:1315-1322. [PMID: 31316623 DOI: 10.3892/etm.2019.7670] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 05/10/2019] [Indexed: 01/12/2023] Open
Abstract
Glioblastoma multiforme is the most aggressive primary brain cancer in adults. Therefore, it is important to investigate the mechanisms associated with cell viability and invasion ability of the cells in glioblastoma multiforme. The opposite strand of the glial cell line-derived neurotrophic factor (GDNF) gene is used to transcribe the cis-antisense GDNF opposite strand (GDNFOS) gene, which belongs to the long noncoding RNAs. The current study assessed the effects of GDNFOS1 overexpression and interference on GDNF expression, cell viability and invasion ability in U87 and U251 MG glioblastoma cells. Overexpression and interference were performed using constructed lentiviral vectors, including long non-coding RNA GDNFOS1 overexpression vector, pL-short hairpin RNA (shRNA)-GDNFOS1-9, pL-shRNA-GDNFOS1-49, pL-shRNA-GDNFOS1-248, pL-shRNA-GDNFOS1-9+49, pL-shRNA-GDNFOS1-9+248 and pL-shRNA-GDNFOS1-49+248. Reverse transcription-quantitative PCR was used to determine the efficiency of interference and overexpression of GDNFOS1 in U87 and U251 MG cells. GDNF protein expression in U87 and U251 MG cells was detected using western blot analysis. In addition, cell viability was detected using a cell counting kit-8 assay at 24, 48 and 72 h after GDNFOS1 overexpression or interference. A transwell invasion assay was used to detect invasion ability. Different shRNA sequences were tested and the results revealed that a combination (pL-shRNA-GDNFOS1-49+248) was most effective in the knock-down GDNFOS1. Compared with the control group, GDNF expression in U87 MG cells was significantly increased in the GDNFOS1 overexpression group and decreased in the shRNA-GDNFOS1-248 group. U87 MG cell viability was significantly increased in the GDNFOS1 overexpression group at 24, 48 and 72 h compared with the negative control group. The viability of U87 MG cells was decreased in the GDNFOS1 interference group at 72 h when compared with the control group. The relative invasive ability was significantly increased in the GDNFOS1 overexpression group when compared with the negative control group. The invasive ability was significantly decreased in the GDNFOS1 interference group when compared with the negative control group. Similar results were exhibited by the U251 MG cells. Overall, GDNF expression, cell viability and invasion ability of glioblastoma cells significantly increased with GDNFOS1 overexpression and decreased with GDNFOS1 interference.
Collapse
Affiliation(s)
- Shiyi Wang
- Department of Gastroenterology, Ningbo Hospital of TCM Affiliated to Zhejiang Chinese Medical University, Ningbo, Zhejiang 315000, P.R. China.,Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, P.R. China
| | - Yihong Fan
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, P.R. China
| | - Yi Xu
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, P.R. China
| | - Lu Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, P.R. China
| | - Lijun Cai
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, P.R. China
| | - Bin Lv
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, P.R. China
| |
Collapse
|
24
|
Xie F, Zhang F, Min S, Chen J, Yang J, Wang X. Glial cell line-derived neurotrophic factor (GDNF) attenuates the peripheral neuromuscular dysfunction without inhibiting the activation of spinal microglia/monocyte. BMC Geriatr 2018; 18:110. [PMID: 29743034 PMCID: PMC5944173 DOI: 10.1186/s12877-018-0796-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Accepted: 04/30/2018] [Indexed: 12/13/2022] Open
Abstract
Background Peripheral neuromuscular dysfunctions were found in elderly individuals, and spinal microglia/monocyte plays an important role on this process. This study aims to test whether the glial cell line-derived neurotrophic factor (GDNF) could attenuate age-related neuromuscular dysfunction by inhibiting the activation of spinal microglia/monocyte. Methods Male Sprague-Dawley rats were divided into an adult group and an aged group. The aged rats were intrathecally injected with normal saline (NS) and GDNF. All the rats were harvested 5 days after each injection. The muscular function was tested by compound muscle action potential, and the activation of microglia/monocyte was detected by immunofluorescence staining; cytokines were assayed by enzyme-linked immunosorbent assay; the expression level of GDNF and its known receptor GFR-α in the spinal cord, the expression level of neuregulin-1 (NRG-1) in the sciatic nerve, and the expression level of γ- and α7- ε-nicotinic acetylcholine receptors in the tibialis anterior muscle were measured by western blotting. Results The activated microglia/monocyte was found in the aged rats compared to the adult rats. The aged rats showed a significant neuromuscular dysfunction and cytokine release as well as increased expression of γ- and α7-nAChR. The protein expression of GDNF, GFR-α, and NRG-1 in the aged rats were significantly lower than that in the adult rats. However, the exogenous injection of GDNF could alleviate the neuromuscular dysfunction but not inhibit the activation of spinal microglia/monocyte. Furthermore, the levels of GFR-α and NRG-1 also increased after GDNF treatment. Conclusion The GDNF could attenuate the age-related peripheral neuromuscular dysfunction without inhibiting the activation of microglia/monocyte in the spinal cord.
Collapse
Affiliation(s)
- Fei Xie
- Department of Anesthesiology, the First Affiliated Hospital of Chongqing Medical University, Friendship Road 1#, Yuan Jia Gang, Chongqing, 400016, China
| | - Fan Zhang
- Department of Anesthesiology, the People's Hospital of Jianyang City, Chengdu, Sichuan, China
| | - Su Min
- Department of Anesthesiology, the First Affiliated Hospital of Chongqing Medical University, Friendship Road 1#, Yuan Jia Gang, Chongqing, 400016, China.
| | - Jingyuan Chen
- Department of Anesthesiology, the First Affiliated Hospital of Chongqing Medical University, Friendship Road 1#, Yuan Jia Gang, Chongqing, 400016, China
| | - Jun Yang
- Department of Anesthesiology, the First Affiliated Hospital of Chongqing Medical University, Friendship Road 1#, Yuan Jia Gang, Chongqing, 400016, China
| | - Xin Wang
- Department of Anesthesiology, the First Affiliated Hospital of Chongqing Medical University, Friendship Road 1#, Yuan Jia Gang, Chongqing, 400016, China
| |
Collapse
|
25
|
GFRA1: A Novel Molecular Target for the Prevention of Osteosarcoma Chemoresistance. Int J Mol Sci 2018; 19:ijms19041078. [PMID: 29617307 PMCID: PMC5979596 DOI: 10.3390/ijms19041078] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/26/2018] [Accepted: 03/31/2018] [Indexed: 12/11/2022] Open
Abstract
The glycosylphosphatidylinositol-linked GDNF (glial cell derived neurotrophic factor) receptor alpha (GFRA), a coreceptor that recognizes the GDNF family of ligands, has a crucial role in the development and maintenance of the nervous system. Of the four identified GFRA isoforms, GFRA1 specifically recognizes GDNF and is involved in the regulation of proliferation, differentiation, and migration of neuronal cells. GFRA1 has also been implicated in cancer cell progression and metastasis. Recent findings show that GFRA1 can contribute to the development of chemoresistance in osteosarcoma. GFRA1 expression was induced following treatment of osteosarcoma cells with the popular anticancer drug, cisplatin and induction of GFRA1 expression significantly suppressed apoptosis mediated by cisplatin in osteosarcoma cells. GFRA1 expression promotes autophagy by activating the SRC-AMPK signaling axis following cisplatin treatment, resulting in enhanced osteosarcoma cell survival. GFRA1-induced autophagy promoted tumor growth in mouse xenograft models, suggesting a novel function of GFRA1 in osteosarcoma chemoresistance.
Collapse
|
26
|
Rosich K, Hanna BF, Ibrahim RK, Hellenbrand DJ, Hanna A. The Effects of Glial Cell Line-Derived Neurotrophic Factor after Spinal Cord Injury. J Neurotrauma 2017; 34:3311-3325. [DOI: 10.1089/neu.2017.5175] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Affiliation(s)
- Konstantin Rosich
- Department of Neurological Surgery, University of Wisconsin, Madison, Wisconsin
| | - Bishoy F. Hanna
- Department of Neurological Surgery, Ross University School of Medicine, Dominica, West Indies
| | - Rami K. Ibrahim
- Department of Neurological Surgery, University of Wisconsin, Madison, Wisconsin
| | - Daniel J. Hellenbrand
- Department of Neurological Surgery, University of Wisconsin, Madison, Wisconsin
- Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin
| | - Amgad Hanna
- Department of Neurological Surgery, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|
27
|
Choi DJ, Eun JH, Kim BG, Jou I, Park SM, Joe EH. A Parkinson's disease gene, DJ-1, repairs brain injury through Sox9 stabilization and astrogliosis. Glia 2017; 66:445-458. [PMID: 29105838 DOI: 10.1002/glia.23258] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 10/23/2017] [Accepted: 10/24/2017] [Indexed: 12/13/2022]
Abstract
Defects in repair of damaged brain accumulate injury and contribute to slow-developing neurodegeneration. Here, we report that a deficiency of DJ-1, a Parkinson's disease (PD) gene, delays repair of brain injury due to destabilization of Sox9, a positive regulator of astrogliosis. Stereotaxic injection of ATP into the brain striatum produces similar size of acute injury in wild-type and DJ-1-knockout (KO) mice. However, recovery of the injury is delayed in KO mice, which is confirmed by 9.4T magnetic resonance imaging and tyrosine hydroxylase immunostaining. DJ-1 regulates neurite outgrowth from damaged neurons in a non-cell autonomous manner. In DJ-1 KO brains and astrocytes, Sox9 protein levels are decreased due to enhanced ubiquitination, resulting in defects in astrogliosis and glial cell-derived neurotrophic factor/ brain-derived neurotrophic factor expression in injured brain and astrocytes. These results indicate that DJ-1 deficiency causes defects in astrocyte-mediated repair of brain damage, which may contribute to the development of PD.
Collapse
Affiliation(s)
- Dong-Joo Choi
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Department of Pharmacology, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea
| | - Jin-Hwa Eun
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Department of Pharmacology, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea
| | - Byung Gon Kim
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Department of Brain Science, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Department of Neurology, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea
| | - Ilo Jou
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Department of Pharmacology, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea
| | - Sang Myun Park
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Department of Pharmacology, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea
| | - Eun-Hye Joe
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Department of Pharmacology, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Department of Brain Science, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea
| |
Collapse
|
28
|
Zaman V, Li Z, Middaugh L, Ramamoorthy S, Rohrer B, Nelson ME, Tomac AC, Hoffer BJ, Gerhardt GA, Granholm AC. The Noradrenergic System of Aged GDNF Heterozygous Mice. Cell Transplant 2017; 12:291-303. [DOI: 10.3727/000000003108746740] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) is a trophic factor for noradrenergic (NE) neurons of the pontine nucleus locus coeruleus (LC). Decreased function of the LC-NE neurons has been found during normal aging and in neurodegenerative disorders. We have previously shown that GDNF participates in the differentiation of LC-NE neurons during development. However, the continued role of GDNF for LC-NE neurons during maturation and aging has not been addressed. We examined alterations in aged mice that were heterozygous for the GDNF gene (Gdnf+/–). Wild-type (Gdnf+/+) and Gdnf+/– mice (18 months old) were tested for locomotor activity and brain tissues were collected for measuring norepinephrine levels and uptake, as well as for morphological analysis. Spontaneous locomotion was reduced in Gdnf+/– mice in comparison with Gdnf+/+ mice. The reduced locomotor activity of Gdnf +/– mice was accompanied by reductions in NE transporter activity in the cerebellum and brain stem as well as decreased norepinephrine tissue levels in the LC. Tyrosine hydroxylase (TH) immunostaining demonstrated morphological alterations of LC-NE cell bodies and abnormal TH-positive fibers in the hippocampus, cerebellum, and frontal cortex of Gdnf+/– mice. These findings suggest that the LC-NE system of Gdnf+/– mice is impaired and suggest that GDNF plays an important role in continued maintenance of this neuronal system throughout life.
Collapse
Affiliation(s)
- V. Zaman
- Department of Physiology and Neuroscience and the Center on Aging, Medical University of South Carolina, Charleston, SC 29425
| | - Z. Li
- Department of Neurology, Medical University of South Carolina, Charleston, SC 29425
| | - L. Middaugh
- Department of Psychiatry, Medical University of South Carolina, Charleston, SC 29425
| | - S. Ramamoorthy
- Department of Physiology and Neuroscience and the Center on Aging, Medical University of South Carolina, Charleston, SC 29425
| | - B. Rohrer
- Department of Physiology and Neuroscience and the Center on Aging, Medical University of South Carolina, Charleston, SC 29425
- Department of Ophthalmology, Medical University of South Carolina, Charleston, SC 29425
| | - M. E. Nelson
- Department of Physiology and Neuroscience and the Center on Aging, Medical University of South Carolina, Charleston, SC 29425
| | - A. C. Tomac
- National Institute on Drug Abuse, IRP, Baltimore, MD 21224
| | - B. J. Hoffer
- National Institute on Drug Abuse, IRP, Baltimore, MD 21224
| | - G. A. Gerhardt
- Chandler Medical Center, Department of Anatomy & Neurobiology, University of Kentucky, Lexington, KY 40536-0098
| | - A. Ch. Granholm
- Department of Physiology and Neuroscience and the Center on Aging, Medical University of South Carolina, Charleston, SC 29425
| |
Collapse
|
29
|
Park HJ, Bolton EC. RET-mediated glial cell line-derived neurotrophic factor signaling inhibits mouse prostate development. Development 2017; 144:2282-2293. [PMID: 28506996 DOI: 10.1242/dev.145086] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 05/10/2017] [Indexed: 01/15/2023]
Abstract
In humans and rodents, the prostate gland develops from the embryonic urogenital sinus (UGS). The androgen receptor (AR) is thought to control the expression of morphogenetic genes in inductive UGS mesenchyme, which promotes proliferation and cytodifferentiation of the prostatic epithelium. However, the nature of the AR-regulated morphogenetic genes and the mechanisms whereby AR controls prostate development are not understood. Glial cell line-derived neurotrophic factor (GDNF) binds GDNF family receptor α1 (GFRα1) and signals through activation of RET tyrosine kinase. Gene disruption studies in mice have revealed essential roles for GDNF signaling in development; however, its role in prostate development is unexplored. Here, we establish novel roles of GDNF signaling in mouse prostate development. Using an organ culture system for prostate development and Ret mutant mice, we demonstrate that RET-mediated GDNF signaling in UGS increases proliferation of mesenchyme cells and suppresses androgen-induced proliferation and differentiation of prostate epithelial cells, inhibiting prostate development. We also identify Ar as a GDNF-repressed gene and Gdnf and Gfrα1 as androgen-repressed genes in UGS, thus establishing reciprocal regulatory crosstalk between AR and GDNF signaling in prostate development.
Collapse
Affiliation(s)
- Hyun-Jung Park
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Eric C Bolton
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
30
|
Tsybko AS, Ilchibaeva TV, Popova NK. Role of glial cell line-derived neurotrophic factor in the pathogenesis and treatment of mood disorders. Rev Neurosci 2017; 28:219-233. [DOI: 10.1515/revneuro-2016-0063] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 11/03/2016] [Indexed: 12/31/2022]
Abstract
AbstractGlial cell line-derived neurotrophic factor (GDNF) is widely recognized as a survival factor for dopaminergic neurons, but GDNF has also been shown to promote development, differentiation, and protection of other central nervous system neurons and was thought to play an important role in various neuropsychiatric disorders. Severe mood disorders, such as primarily major depressive disorder and bipolar affective disorder, attract particular attention. These psychopathologies are characterized by structural alterations accompanied by the dysregulation of neuroprotective and neurotrophic signaling mechanisms required for the maturation, growth, and survival of neurons and glia. The main objective of this review is to summarize the recent findings and evaluate the potential role of GDNF in the pathogenesis and treatment of mood disorders. Specifically, it describes (1) the implication of GDNF in the mechanism of depression and in the effect of antidepressant drugs and mood stabilizers and (2) the interrelation between GDNF and brain neurotransmitters, playing a key role in the pathogenesis of depression. This review provides converging lines of evidence that (1) brain GDNF contributes to the mechanism underlying depressive disorders and the effect of antidepressants and mood stabilizers and (2) there is a cross-talk between GDNF and neurotransmitters representing a feedback system: GDNF-neurotransmitters and neurotransmitters-GDNF.
Collapse
Affiliation(s)
- Anton S. Tsybko
- 1Department of Behavioral Neurogenomics, The Federal Research Center the Institute of Cytology and Genetics SB RAS, Lavrentyeva av. 10, Novosibirsk 630090, Russia
| | - Tatiana V. Ilchibaeva
- 2Department of Behavioral Neurogenomics, The Federal Research Center the Institute of Cytology and Genetics SB RAS, Novosibirsk 633090, Russia
| | - Nina K. Popova
- 2Department of Behavioral Neurogenomics, The Federal Research Center the Institute of Cytology and Genetics SB RAS, Novosibirsk 633090, Russia
| |
Collapse
|
31
|
Yue P, Gao L, Wang X, Ding X, Teng J. Intranasal Administration of GDNF Protects Against Neural Apoptosis in a Rat Model of Parkinson’s Disease Through PI3K/Akt/GSK3β Pathway. Neurochem Res 2017; 42:1366-1374. [DOI: 10.1007/s11064-017-2184-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 01/13/2017] [Accepted: 01/17/2017] [Indexed: 12/16/2022]
|
32
|
Ibáñez CF, Andressoo JO. Biology of GDNF and its receptors — Relevance for disorders of the central nervous system. Neurobiol Dis 2017; 97:80-89. [DOI: 10.1016/j.nbd.2016.01.021] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 01/14/2016] [Accepted: 01/25/2016] [Indexed: 01/15/2023] Open
|
33
|
Corrigan F, Arulsamy A, Teng J, Collins-Praino LE. Pumping the Brakes: Neurotrophic Factors for the Prevention of Cognitive Impairment and Dementia after Traumatic Brain Injury. J Neurotrauma 2016; 34:971-986. [PMID: 27630018 DOI: 10.1089/neu.2016.4589] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) is the leading cause of disability and death worldwide, affecting as many as 54,000,000-60,000,000 people annually. TBI is associated with significant impairments in brain function, impacting cognitive, emotional, behavioral, and physical functioning. Although much previous research has focused on the impairment immediately following injury, TBI may have much longer-lasting consequences, including neuropsychiatric disorders and cognitive impairment. TBI, even mild brain injury, has also been recognized as a significant risk factor for the later development of dementia and Alzheimer's disease. Although the link between TBI and dementia is currently unknown, several proposed mechanisms have been put forward, including alterations in glucose metabolism, excitotoxicity, calcium influx, mitochondrial dysfunction, oxidative stress, and neuroinflammation. A treatment for the devastating long-term consequences of TBI is desperately needed. Unfortunately, however, no such treatment is currently available, making this a major area of unmet medical need. Increasing the level of neurotrophic factor expression in key brain areas may be one potential therapeutic strategy. Of the neurotrophic factors, granulocyte-colony stimulating factor (G-CSF) may be particularly effective for preventing the emergence of long-term complications of TBI, including dementia, because of its ability to reduce apoptosis, stimulate neurogenesis, and increase neuroplasticity.
Collapse
Affiliation(s)
- Frances Corrigan
- Translational Neuropathology Lab, Discipline of Anatomy and Pathology, School of Medicine, University of Adelaide , Adelaide, Australia
| | - Alina Arulsamy
- Translational Neuropathology Lab, Discipline of Anatomy and Pathology, School of Medicine, University of Adelaide , Adelaide, Australia
| | - Jason Teng
- Translational Neuropathology Lab, Discipline of Anatomy and Pathology, School of Medicine, University of Adelaide , Adelaide, Australia
| | - Lyndsey E Collins-Praino
- Translational Neuropathology Lab, Discipline of Anatomy and Pathology, School of Medicine, University of Adelaide , Adelaide, Australia
| |
Collapse
|
34
|
Gabreski NA, Vaghasia JK, Novakova SS, McDonald NQ, Pierchala BA. Exon Skipping in the RET Gene Encodes Novel Isoforms That Differentially Regulate RET Protein Signal Transduction. J Biol Chem 2016; 291:16249-62. [PMID: 27226544 PMCID: PMC4965573 DOI: 10.1074/jbc.m115.709675] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 05/09/2016] [Indexed: 01/15/2023] Open
Abstract
Rearranged during transfection (RET), a receptor tyrosine kinase that is activated by the glial cell line-derived neurotrophic factor family ligands (GFLs), plays a crucial role in the development and function of the nervous system and additionally is required for kidney development and spermatogenesis. RET encodes a transmembrane receptor that is 20 exons long and produces two known protein isoforms differing in C-terminal amino acid composition, referred to as RET9 and RET51. Studies of human pheochromocytomas identified two additional novel transcripts involving the skipping of exon 3 or exons 3, 4, and 5 and are referred to as RET(Δ) (E3) and RET(Δ) (E345), respectively. Here we report the presence of Ret(Δ) (E3) and Ret(Δ) (E345) in zebrafish, mice, and rats and show that these transcripts are dynamically expressed throughout development of the CNS, peripheral nervous system, and kidneys. We further explore the biochemical properties of these isoforms, demonstrating that, like full-length RET, RET(ΔE3) and RET(ΔE345) are trafficked to the cell surface, interact with all four GFRα co-receptors, and have the ability to heterodimerize with full-length RET. Signaling experiments indicate that RET(ΔE3) is phosphorylated in a similar manner to full-length RET. RET(ΔE345), in contrast, displays higher baseline autophosphorylation, specifically on the catalytic tyrosine, Tyr(905), and also on one of the most important signaling residues, Tyr(1062) These data provide the first evidence for a physiologic role of these isoforms in RET pathway function.
Collapse
Affiliation(s)
- Nicole A Gabreski
- From the Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, Michigan 48109, the Program in Cellular and Molecular Biology, University of Michigan School of Medicine, Ann Arbor, Michigan 48109
| | - Janki K Vaghasia
- From the Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, Michigan 48109
| | - Silvia S Novakova
- From the Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, Michigan 48109
| | - Neil Q McDonald
- the Structural Biology Laboratory, Francis Crick Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, United Kingdom, and the Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck College, Malet Street, London WC1E 7HX, United Kingdom
| | - Brian A Pierchala
- From the Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, Michigan 48109, the Program in Cellular and Molecular Biology, University of Michigan School of Medicine, Ann Arbor, Michigan 48109,
| |
Collapse
|
35
|
Gao YY, Hong XY, Wang HJ. Role of Nectin-1/c-Src Signaling in the Analgesic Effect of GDNF on a Rat Model of Chronic Constrictive Injury. J Mol Neurosci 2016; 60:258-66. [DOI: 10.1007/s12031-016-0792-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 06/30/2016] [Indexed: 12/20/2022]
|
36
|
Kramer ER, Liss B. GDNF-Ret signaling in midbrain dopaminergic neurons and its implication for Parkinson disease. FEBS Lett 2015; 589:3760-72. [DOI: 10.1016/j.febslet.2015.11.006] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 10/29/2015] [Accepted: 11/03/2015] [Indexed: 12/11/2022]
|
37
|
Budni J, Bellettini-Santos T, Mina F, Garcez ML, Zugno AI. The involvement of BDNF, NGF and GDNF in aging and Alzheimer's disease. Aging Dis 2015; 6:331-41. [PMID: 26425388 DOI: 10.14336/ad.2015.0825] [Citation(s) in RCA: 280] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 08/25/2015] [Indexed: 12/21/2022] Open
Abstract
Aging is a normal physiological process accompanied by cognitive decline. This aging process has been the primary risk factor for development of aging-related diseases such as Alzheimer's disease (AD). Cognitive deficit is related to alterations of neurotrophic factors level such as brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF) and glial cell-derived neurotrophic factor (GDNF). These strong relationship between aging and AD is important to investigate the time which they overlap, as well as, the pathophysiological mechanism in each event. Considering that aging and AD are related to cognitive impairment, here we discuss the involving these neurotrophic factors in the aging process and AD.
Collapse
Affiliation(s)
- Josiane Budni
- Laboratório de Neurociências, Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Tatiani Bellettini-Santos
- Laboratório de Neurociências, Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Francielle Mina
- Laboratório de Neurociências, Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Michelle Lima Garcez
- Laboratório de Neurociências, Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Alexandra Ioppi Zugno
- Laboratório de Neurociências, Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| |
Collapse
|
38
|
Janssen S, Schlegel C, Gudi V, Prajeeth CK, Skripuletz T, Trebst C, Stangel M. Effect of FTY720-phosphate on the expression of inflammation-associated molecules in astrocytes in vitro. Mol Med Rep 2015; 12:6171-7. [PMID: 26239526 DOI: 10.3892/mmr.2015.4120] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 05/21/2015] [Indexed: 11/06/2022] Open
Abstract
FTY720 is a new oral immunomodulatory therapy for the treatment of multiple sclerosis (MS). There is strong evidence that FTY720 has direct effects on brain resident cells such as astrocytes acting via sphingosine‑1‑phosphate (S1P) receptors. In the present study, the mRNA expression of S1P receptors as well as selected cytokines, chemokines and growth factors were investigated in primary murine astrocytes under inflammatory conditions in the presence or absence of the phosphorylated form of FTY720 (FTY720‑P). Following stimulation with either the pro‑inflammatory cytokine tumor necrosis factor‑α (TNF‑α) or with bacterial lipopolysaccharide, there was an increased expression of the receptors S1P1 and S1P3, the cytokines and chemokines interleukin (IL)‑1β, chemokine (C‑C‑motif) ligand 2 (CCL‑2), CCL‑20 and chemokine (C‑X‑C‑motif) ligand 12 as well as the growth factors insulin‑like growth factor‑1, ciliary neurotrophic factor and glial cell line‑derived neurotrophic factor (GDNF). FTY720‑P led to an increased expression of IL‑1β and GDNF at distinct time points following co‑stimulation with TNF‑α compared with TNF‑α treatment alone. However, the presence of FTY720‑P did not have any further significant effects on the expression of S1P receptors, cytokines or growth factors, suggesting that the regulation of these target genes in astrocytes is not likely to be a major mechanism underlying the effect of FTY720‑P in diseases such as MS.
Collapse
Affiliation(s)
- Stefanie Janssen
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Hannover D‑30625, Germany
| | - Caroline Schlegel
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Hannover D‑30625, Germany
| | - Viktoria Gudi
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Hannover D‑30625, Germany
| | - Chittappen Kandiyil Prajeeth
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Hannover D‑30625, Germany
| | - Thomas Skripuletz
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Hannover D‑30625, Germany
| | - Corinna Trebst
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Hannover D‑30625, Germany
| | - Martin Stangel
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Hannover D‑30625, Germany
| |
Collapse
|
39
|
Abstract
ABSTRACT
Midbrain dopaminergic (mDA) neuron development has been an intense area of research during recent years. This is due in part to a growing interest in regenerative medicine and the hope that treatment for diseases affecting mDA neurons, such as Parkinson's disease (PD), might be facilitated by a better understanding of how these neurons are specified, differentiated and maintained in vivo. This knowledge might help to instruct efforts to generate mDA neurons in vitro, which holds promise not only for cell replacement therapy, but also for disease modeling and drug discovery. In this Primer, we will focus on recent developments in understanding the molecular mechanisms that regulate the development of mDA neurons in vivo, and how they have been used to generate human mDA neurons in vitro from pluripotent stem cells or from somatic cells via direct reprogramming. Current challenges and future avenues in the development of a regenerative medicine for PD will be identified and discussed.
Collapse
Affiliation(s)
- Ernest Arenas
- Laboratory of Molecular Neurobiology, Dept. Medical Biochemistry and Biophysics, Center of Developmental Biology for Regenerative Medicine, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Mark Denham
- Laboratory of Molecular Neurobiology, Dept. Medical Biochemistry and Biophysics, Center of Developmental Biology for Regenerative Medicine, Karolinska Institutet, Stockholm 171 77, Sweden
- Danish Research Institute of Translational Neuroscience, Nordic EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus 8000, Denmark
| | - J. Carlos Villaescusa
- Laboratory of Molecular Neurobiology, Dept. Medical Biochemistry and Biophysics, Center of Developmental Biology for Regenerative Medicine, Karolinska Institutet, Stockholm 171 77, Sweden
- Institute of Experimental Biology, Faculty of Science, Masaryk University, Brno 61137, Czech Republic
| |
Collapse
|
40
|
Kopra J, Vilenius C, Grealish S, Härma MA, Varendi K, Lindholm J, Castrén E, Võikar V, Björklund A, Piepponen TP, Saarma M, Andressoo JO. GDNF is not required for catecholaminergic neuron survival in vivo. Nat Neurosci 2015; 18:319-22. [PMID: 25710828 DOI: 10.1038/nn.3941] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Jaakko Kopra
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Carolina Vilenius
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Shane Grealish
- Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Mari-Anne Härma
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Kärt Varendi
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Jesse Lindholm
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Eero Castrén
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Vootele Võikar
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Anders Björklund
- Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - T Petteri Piepponen
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Mart Saarma
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | | |
Collapse
|
41
|
Kimura M, Sakai A, Sakamoto A, Suzuki H. Glial cell line-derived neurotrophic factor-mediated enhancement of noradrenergic descending inhibition in the locus coeruleus exerts prolonged analgesia in neuropathic pain. Br J Pharmacol 2015; 172:2469-78. [PMID: 25572945 DOI: 10.1111/bph.13073] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 10/10/2014] [Accepted: 12/25/2014] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND AND PURPOSE The locus coeruleus (LC) is the principal nucleus containing the noradrenergic neurons and is a major endogenous source of pain modulation in the brain. Glial cell line-derived neurotrophic factor (GDNF), a well-established neurotrophic factor for noradrenergic neurons, is a major pain modulator in the spinal cord and primary sensory neurons. However, it is unknown whether GDNF is involved in pain modulation in the LC. EXPERIMENTAL APPROACH Rats with chronic constriction injury (CCI) of the left sciatic nerve were used as a model of neuropathic pain. GDNF was injected into the left LC of rats with CCI for 3 consecutive days and changes in mechanical allodynia and thermal hyperalgesia were assessed. The α2 -adrenoceptor antagonist yohimbine was injected intrathecally to assess the involvement of descending inhibition in GDNF-mediated analgesia. The MEK inhibitor U0126 was used to investigate whether the ERK signalling pathway plays a role in the analgesic effects of GDNF. KEY RESULTS Both mechanical allodynia and thermal hyperalgesia were attenuated 24 h after the first GDNF injection. GDNF increased the noradrenaline content in the dorsal spinal cord. The analgesic effects continued for at least 3 days after the last injection. Yohimbine abolished these effects of GDNF. The analgesic effects of GDNF were partly, but significantly, inhibited by prior injection of U0126 into the LC. CONCLUSIONS AND IMPLICATIONS GDNF injection into the LC exerts prolonged analgesic effects on neuropathic pain in rats by enhancing descending noradrenergic inhibition.
Collapse
Affiliation(s)
- M Kimura
- Department of Anesthesiology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | | | | | | |
Collapse
|
42
|
Razavi S, Nazem G, Mardani M, Esfandiari E, Salehi H, Esfahani SHZ. Neurotrophic factors and their effects in the treatment of multiple sclerosis. Adv Biomed Res 2015; 4:53. [PMID: 25802822 PMCID: PMC4361963 DOI: 10.4103/2277-9175.151570] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 04/24/2014] [Indexed: 12/24/2022] Open
Abstract
Neurotrophins are small molecules of polypeptides, which include nerve growth factor (NGF) family, glial cell line–derived neurotrophic factor (GDNF) family ligands, and neuropoietic cytokines. These factors have an important role in neural regeneration, remyelination, and regulating the development of the peripheral and central nervous systems (PNS and CNS, respectively) by intracellular signaling through specific receptors. It has been suggested that the pathogenesis of human neurodegenerative disorders may be due to an alteration in the neurotrophic factors and their receptors. The use of neurotrophic factors as therapeutic agents is a novel strategy for restoring and maintaining neuronal function during neurodegenerative disorders such as multiple sclerosis. Innate and adaptive immune responses contribute to pathology of neurodegenerative disorders. Furthermore, autoimmune and mesenchymal stem cells, by the release of neurotrophic factors, have the ability to protect neuronal population and can efficiently suppress the formation of new lesions. So, these cells may be an alternative source for delivering neurotrophic factors into the CNS.
Collapse
Affiliation(s)
- Shahnaz Razavi
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ghasemi Nazem
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Mardani
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ebrahim Esfandiari
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Salehi
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | |
Collapse
|
43
|
Duan W, Zhang YP, Hou Z, Huang C, Zhu H, Zhang CQ, Yin Q. Novel Insights into NeuN: from Neuronal Marker to Splicing Regulator. Mol Neurobiol 2015; 53:1637-1647. [PMID: 25680637 DOI: 10.1007/s12035-015-9122-5] [Citation(s) in RCA: 194] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 02/01/2015] [Indexed: 01/07/2023]
Abstract
Neuronal nuclei (NeuN) is a well-recognized "marker" that is detected exclusively in post-mitotic neurons and was initially identified through an immunological screen to produce neuron-specific antibodies. Immunostaining evidence indicates that NeuN is distributed in the nuclei of mature neurons in nearly all parts of the vertebrate nervous system. NeuN is highly conserved among species and is stably expressed during specific stages of development. Therefore, NeuN has been considered to be a reliable marker of mature neurons for the past two decades. However, this role has been challenged by recent studies indicating that NeuN staining is variable and even absent during certain diseases and specific physiological states. More importantly, despite the widespread use of the anti-NeuN antibody, the natural identity of the NeuN protein remained elusive for 17 years. NeuN was recently eventually identified as an epitope of Rbfox3, which is a novel member of the Rbfox1 family of splicing factors. This identification might provide a novel perspective on NeuN expression during both physiological and pathological conditions. This review summarizes the current progress on the biochemical identity and biological significance of NeuN and recommends caution when applying NeuN immunoreactivity as a definitive marker of mature neurons in certain diseases and specific physiological states.
Collapse
Affiliation(s)
- Wei Duan
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, People's Republic of China
| | - Yu-Ping Zhang
- Department of Pediatrics, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, People's Republic of China
| | - Zhi Hou
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, People's Republic of China
| | - Chen Huang
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, 37232, USA
| | - He Zhu
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, 37232, USA.,Department of Biological Sciences, Vanderbilt University, Nashville, TN, 37232, USA.,Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA.,The Vanderbilt Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN, 37232, USA
| | - Chun-Qing Zhang
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, People's Republic of China.
| | - Qing Yin
- Department of Rehabilitation and Physical Therapy, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, People's Republic of China.
| |
Collapse
|
44
|
Ghasemi N. Therapeutic effects of adipose derived mesenchymal stem cells on remyelination process in inflammatory demyelinating diseases. ACTA ACUST UNITED AC 2015. [DOI: 10.7243/2055-091x-2-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
45
|
Park HJ, Bolton EC. Glial cell line-derived neurotrophic factor induces cell proliferation in the mouse urogenital sinus. Mol Endocrinol 2014; 29:289-306. [PMID: 25549043 DOI: 10.1210/me.2014-1312] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) is a TGFβ family member, and GDNF signals through a glycosyl-phosphatidylinositol-linked cell surface receptor (GFRα1) and RET receptor tyrosine kinase. GDNF signaling plays crucial roles in urogenital processes, ranging from cell fate decisions in germline progenitors to ureteric bud outgrowth and renal branching morphogenesis. Gene ablation studies in mice have revealed essential roles for GDNF signaling in urogenital development, although its role in prostate development is unclear. We investigated the functional role of GDNF signaling in the urogenital sinus (UGS) and the developing prostate of mice. GDNF, GFRα1, and RET show time-specific and cell-specific expression during prostate development in vivo. In the UGS, GDNF and GFRα1 are expressed in the urethral mesenchyme (UrM) and epithelium (UrE), whereas RET is restricted to the UrM. In each lobe of the developing prostate, GDNF and GFRα1 expression declines in the epithelium and becomes restricted to the stroma. Using a well-established organ culture system, we determined that exogenous GDNF increases proliferation of UrM and UrE cells, altering UGS morphology. With regard to mechanism, GDNF signaling in the UrM increased RET expression and phosphorylation of ERK1/2. Furthermore, inhibition of RET kinase activity or ERK kinases suppressed GDNF-induced proliferation of UrM cells but not UrE cells. We therefore propose that GDNF signaling in the UGS increases proliferation of UrM and UrE cells by different mechanisms, which are distinguished by the role of RET receptor tyrosine kinase and ERK kinase signaling, thus implicating GDNF signaling in prostate development and growth.
Collapse
Affiliation(s)
- Hyun-Jung Park
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801
| | | |
Collapse
|
46
|
Broom L, Jenner P, Rose S. Increased neurotrophic factor levels in ventral mesencephalic cultures do not explain the protective effect of osteopontin and the synthetic 15-mer RGD domain against MPP+ toxicity. Exp Neurol 2014; 263:1-7. [PMID: 25218309 DOI: 10.1016/j.expneurol.2014.09.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2014] [Revised: 09/02/2014] [Accepted: 09/05/2014] [Indexed: 01/13/2023]
Abstract
The synthetic 15-mer arginine-glycine-aspartic acid (RGD) domain of osteopontin (OPN) is protective in vitro and in vivo against dopaminergic cell death and this protective effect may be mediated through interaction with integrin receptors to regulate neurotrophic factor levels. We now examine this concept in rat primary ventral mesencephalic (VM) cultures. 1-Methyl-4-phenylpyridinium (MPP+) exposure reduced tyrosine hydroxylase (TH)-positive cell number and activated glial cells as shown by increased glial fibrillary acidic protein (GFAP), oxycocin-42 (OX-42) and ectodermal dysplasia 1 (ED-1) immunoreactivity. Both OPN and the RGD domain of OPN were equally protective against MPP+ toxicity in VM cultures and both increased glial-derived neurotrophic factor (GDNF) and brain-derived neurotrophic factor (BDNF) levels. The effects of OPN and the RGD domain were accompanied by a decrease in numbers of activated microglia but with no change in astrocyte number. However, full-length OPN and the RGD domain of OPN remained protective against MPP+ toxicity in the presence of a GDNF neutralising antibody. This suggests that increased GDNF levels do not underlie the protective effect observed with OPN. Rather, OPN's protective effect may be mediated through decreased glial cell activation.
Collapse
Affiliation(s)
- Lauren Broom
- Neurodegenerative Diseases Research Group, Institute of Pharmaceutical Science, School of Biomedical Sciences, King's College London, London, SE1 1UL, UK.
| | - Peter Jenner
- Neurodegenerative Diseases Research Group, Institute of Pharmaceutical Science, School of Biomedical Sciences, King's College London, London, SE1 1UL, UK
| | - Sarah Rose
- Neurodegenerative Diseases Research Group, Institute of Pharmaceutical Science, School of Biomedical Sciences, King's College London, London, SE1 1UL, UK
| |
Collapse
|
47
|
Abstract
As stem cells (SCs) in adult organs continue to be identified and characterized, it becomes clear that their survival, quiescence, and activation depend on specific signals in their microenvironment, or niche. Although adult SCs of diverse tissues differ by their developmental origin, cycling activity, and regenerative capacity, there appear to be conserved similarities regarding the cellular and molecular components of the SC niche. Interestingly, many organs house both slow-cycling and fast-cycling SC populations, which rely on the coexistence of quiescent and inductive niches for proper regulation. In this review we present a general definition of adult SC niches in the most studied mammalian systems. We further focus on dissecting their cellular organization and on highlighting recently identified key molecular regulators. Finally, we detail the potential involvement of the SC niche in tissue degeneration, with a particular emphasis on aging and cancer.
Collapse
Affiliation(s)
- Amélie Rezza
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, USA; Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Rachel Sennett
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, USA; Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Michael Rendl
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, USA; Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, USA; Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, USA.
| |
Collapse
|
48
|
Du J, Gao X, Deng L, Chang N, Xiong H, Zheng Y. Transfection of the glial cell line-derived neurotrophic factor gene promotes neuronal differentiation. Neural Regen Res 2014; 9:33-40. [PMID: 25206741 PMCID: PMC4146314 DOI: 10.4103/1673-5374.125327] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2013] [Indexed: 02/01/2023] Open
Abstract
Glial cell line-derived neurotrophic factor recombinant adenovirus vector-transfected bone marrow mesenchymal stem cells were induced to differentiate into neuron-like cells using inductive medium containing retinoic acid and epidermal growth factor. Cell viability, microtubule-associated protein 2-positive cell ratio, and the expression levels of glial cell line-derived neurotrophic factor, nerve growth factor and growth-associated protein-43 protein in the supernatant were significantly higher in glial cell line-derived neurotrophic factor/bone marrow mesenchymal stem cells compared with empty virus plasmid-transfected bone marrow mesenchymal stem cells. Furthermore, microtubule-associated protein 2, glial cell line-derived neurotrophic factor, nerve growth factor and growth-associated protein-43 mRNA levels in cell pellets were statistically higher in glial cell line-derived neurotrophic factor/bone marrow mesenchymal stem cells compared with empty virus plasmid-transfected bone marrow mesenchymal stem cells. These results suggest that glial cell line-derived neurotrophic factor/bone marrow mesenchymal stem cells have a higher rate of induction into neuron-like cells, and this enhanced differentiation into neuron-like cells may be associated with up-regulated expression of glial cell line-derived neurotrophic factor, nerve growth factor and growth-associated protein-43.
Collapse
Affiliation(s)
- Jie Du
- Department of Physiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan Province, China ; Department of Anatomy, Luzhou Medical College, Luzhou, Sichuan Province, China
| | - Xiaoqing Gao
- Research Center for Preclinical Medicine, Luzhou Medical College, Luzhou, Sichuan Province, China
| | - Li Deng
- Research Center for Preclinical Medicine, Luzhou Medical College, Luzhou, Sichuan Province, China
| | - Nengbin Chang
- Department of Anatomy, Luzhou Medical College, Luzhou, Sichuan Province, China
| | - Huailin Xiong
- Department of Anatomy, Luzhou Medical College, Luzhou, Sichuan Province, China
| | - Yu Zheng
- Department of Physiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
49
|
Shilpa J, Paulose CS. GABA and 5-HT chitosan nanoparticles decrease striatal neuronal degeneration and motor deficits during liver injury. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2014; 25:1721-1735. [PMID: 24682906 DOI: 10.1007/s10856-014-5195-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2013] [Accepted: 03/12/2014] [Indexed: 06/03/2023]
Abstract
The metabolic alterations resulted from hepatic injury and cell loss lead to synaptic defects and neurodegeneration that undoubtedly contribute motor deficits. In the present study, GABA and 5-HT chitosan nanoparticles mediated liver cell proliferation influenced by growth factor and cytokines and neuronal survival in corpus striatum of partially hepatectomised rats was evaluated. Liver cell proliferation was initiated and progressed by the combined effect of increased expression of growth factor, insulin like growth factor-1 and decreased expressions of cytokines, tumor necrosis factor-α and Akt-1. This was confirmed by the extent of incorporation of thymidine analogue, BrdU, in the DNA of rapidly dividing cells. Inappropriate influx of compounds to corpus striatum resulting from incomplete metabolism elevated GABAB and 5-HT2A neurotransmissions compared to those treated with nanoparticles. This directly influenced cyclic AMP response element binding protein, glial cell derived neurotrophic factor and brain derived neurotrophic factor in the corpus striatum that facilitate neurogenesis, neuronal survival, development, differentiation and neuroprotection. Motor deficits due to liver injury followed striatal neuronal damage were scored by grid walk and rotarod studies, which confirmed the regain of motor activity by GABA and 5-HT chitosan nanoparticle treatment. The present study revealed the therapeutic significance of GABA and 5-HT chitosan nanoparticles in liver based diseases and related striatal neuronal damage that influenced by GABA and 5-HT.
Collapse
Affiliation(s)
- J Shilpa
- Department of Biotechnology, Molecular Neurobiology and Cell Biology Unit, Centre for Neuroscience, Cochin University of Science and Technology, Cochin, 682 022, Kerala, India
| | | |
Collapse
|
50
|
Immunohistochemical localization of brain-derived neurotrophic factor and glial cell line-derived neurotrophic factor in the superior olivary complex of mice after radiofrequency exposure. Neurosci Lett 2014; 564:78-82. [DOI: 10.1016/j.neulet.2014.02.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 01/22/2014] [Accepted: 02/06/2014] [Indexed: 11/22/2022]
|