1
|
Si J, Yu K, Hao J, Wang J, Zhang L. The therapeutic effects and mechanisms of glucagon-like peptide-1 receptor agonists in neurocognitive disorders. Ther Adv Neurol Disord 2025; 18:17562864251332035. [PMID: 40291753 PMCID: PMC12033604 DOI: 10.1177/17562864251332035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 03/17/2025] [Indexed: 04/30/2025] Open
Abstract
Chronic cerebral hypoperfusion (CCH) represents a key pathogenic contributor to neurocognitive disorders. It can lead to multifaceted pathological alterations including neuroinflammation, neuronal apoptosis, blood-brain barrier disruption, synaptic plasticity deficits, and mitochondrial dysfunction. The glucagon-like peptide-1 receptor (GLP-1R), ubiquitously expressed across multiple organ systems, exerts neuroprotective effects by maintaining intracellular homeostasis and mitigating neuronal damage triggered by oxidative stress, inflammatory cascades, apoptotic signaling, and ischemic insults. Furthermore, GLP-1R activity is modulated by gut microbiota composition and short-chain fatty acid abundance, implicating the gut-brain axis in its regulatory influence on neurological function. This review systematically examines the pathophysiological mechanisms underlying CCH and highlights the therapeutic potential of GLP-1R activation. Specifically, GLP-1R-targeted interventions attenuate hypoperfusion-induced damage through pleiotropic pathways and gut-brain crosstalk, thereby offering novel perspectives for advancing both fundamental research and clinical management of neurocognitive disorders.
Collapse
Affiliation(s)
- Junchen Si
- Department of Neurosurgery, Liaocheng People’s Hospital, Liaocheng, Shandong, China
| | - Kai Yu
- Department of Burn and Plastic Surgery, Liaocheng People’s Hospital, Liaocheng, Shandong, China
| | - Jiheng Hao
- Department of Neurosurgery, Liaocheng People’s Hospital, Liaocheng, Shandong, China
| | - Jiyue Wang
- Department of Neurosurgery, Liaocheng People’s Hospital, Liaocheng, Shandong, China
| | - Liyong Zhang
- Department of Neurosurgery, Liaocheng People’s Hospital, No. 45 Huashan Road, Liaocheng, Shandong 252000, China
| |
Collapse
|
2
|
Pérez-Revuelta L, Pérez-Boyero D, Pérez-Martín E, Cabedo VL, Téllez de Meneses PG, Weruaga E, Díaz D, Alonso JR. Neuroprotective Effects of VEGF-B in a Murine Model of Aggressive Neuronal Loss with Childhood Onset. Int J Mol Sci 2025; 26:538. [PMID: 39859255 PMCID: PMC11765331 DOI: 10.3390/ijms26020538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
In recent decades, the scientific community has faced a major challenge in the search for new therapies that can slow down or alleviate the process of neuronal death that accompanies neurodegenerative diseases. This study aimed to identify an effective therapy using neurotrophic factors to delay the rapid and aggressive cerebellar degeneration experienced by the Purkinje Cell Degeneration (PCD) mouse, a model of childhood-onset neurodegeneration with cerebellar atrophy (CONDCA). Initially, we analyzed the changes in the expression of several neurotrophic factors related to the degenerative process itself, identifying changes in insulin-like growth factor 1 (IGF-1) and Vascular Endothelial Growth Factor B (VEGF-B) in the affected animals. Then, we administered pharmacological treatments using human recombinant IGF-1 (rhIGF-1) or VEGF-B (rhVEGF-B) proteins, considering their temporal variations during the degenerative process. The effects of these treatments on motor, cognitive, and social behavior, as well as on cerebellar destructuration were analyzed. Whereas treatment with rhIGF-1 did not demonstrate any neuroprotective effect, rhVEGF-B administration at moderate dosages stopped the process of neuronal death and restored motor, cognitive, and social functions altered in PCD mice (and CONDCA patients). However, increasing the frequency of rhVEGF-B administration had a detrimental effect on Purkinje cell survival, suggesting an inverted U-shaped dose-response curve of this substance. Additionally, we demonstrate that this neuroprotective effect was achieved through a partial inhibition or delay of apoptosis. These findings provide strong evidence supporting the use of rhVEGF-B as a pharmacological agent to limit severe cerebellar neurodegenerative processes.
Collapse
Affiliation(s)
- Laura Pérez-Revuelta
- Laboratory of Neuronal Plasticity and Neurorepair, Institute of Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, 37007 Salamanca, Spain; (L.P.-R.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - David Pérez-Boyero
- Laboratory of Neuronal Plasticity and Neurorepair, Institute of Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, 37007 Salamanca, Spain; (L.P.-R.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Ester Pérez-Martín
- Neuroscience Innovative Technologies, Neurostech, 33428 Llanera, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Intervenciones Traslacionales para la Salud, 33011 Oviedo, Spain
| | - Valeria Lorena Cabedo
- Laboratory of Neuronal Plasticity and Neurorepair, Institute of Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, 37007 Salamanca, Spain; (L.P.-R.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Pablo González Téllez de Meneses
- Laboratory of Neuronal Plasticity and Neurorepair, Institute of Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, 37007 Salamanca, Spain; (L.P.-R.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Eduardo Weruaga
- Laboratory of Neuronal Plasticity and Neurorepair, Institute of Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, 37007 Salamanca, Spain; (L.P.-R.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - David Díaz
- Laboratory of Neuronal Plasticity and Neurorepair, Institute of Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, 37007 Salamanca, Spain; (L.P.-R.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - José Ramón Alonso
- Laboratory of Neuronal Plasticity and Neurorepair, Institute of Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, 37007 Salamanca, Spain; (L.P.-R.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| |
Collapse
|
3
|
Shandra O, Wang Y, Coles LD, Mowrey WB, Li Q, Liu W, Moshé SL, Galanopoulou AS. Efficacy and tolerability of celastrol and edaravone in the multiple-hit rat model of infantile spasms. Epilepsy Behav 2025; 162:110159. [PMID: 39577370 PMCID: PMC11681605 DOI: 10.1016/j.yebeh.2024.110159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/09/2024] [Accepted: 11/14/2024] [Indexed: 11/24/2024]
Abstract
OBJECTIVE To test whether anti-inflammatory and antioxidant drugs that inhibit the nuclear factor kappa light chain enhancer of activated B cells (NF-kB), celastrol and edaravone, suppress spasms and improve developmental outcomes in the multiple-hit rat model of refractory infantile spasms (IS) due to structural lesions. METHODS Postnatal day 3 (PN3) Sprague-Dawley rats were treated according to the multiple-hit IS model protocol. Using a randomized, blinded, vehicle-controlled, dose- and time-response study design, we tested the effects of single celastrol [1, 2, or 4 mg/kg intraperitoneally (i.p.), 10-14 rats/group] or edaravone (1, 10 or 30 mg/kg i.p., 14-17 rats/group) injections vs their vehicles on behavioral and electroclinical spasms and developmental milestones. Video-EEG monitoring was done on PN6-7 (n = 11-12 rats/group). Pulse celastrol treatment effects (PN4: 4 mg/kg, PN5-6: 2 mg/kg/day i.p.) were determined on spasms, developmental milestones and Barnes maze. Celastrol and edaravone pharmacokinetics in plasma and neocortex were assessed. Linear mixed model analysis of raw or normalized log-transformed spasm frequencies, considering repeated observations was used. RESULTS Single (2-4 mg/kg i.p) or pulse celastrol, but not edaravone, reduced behavioral and electroclinical spasms frequencies within 5hrs. Pulse celastrol did not affect spasm-freedom, survival, developmental milestones or Barnes maze performance. Celastrol had erratic i.p. absorption with maximum concentrations observed between 2-4 h, when effects on spasms were seen. Edaravone had low blood-to-brain permeability. CONCLUSIONS Celastrol's efficacy on spasms is partially explained by its better brain penetration than edaravone's. NFkB inhibitors may be useful in treating drug-resistant IS but delivery methods with improved bioavailability and brain permeability are needed.
Collapse
Affiliation(s)
- Oleksii Shandra
- Albert Einstein College of Medicine, Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Bronx, NY, USA
| | - Yongjun Wang
- Albert Einstein College of Medicine, Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Bronx, NY, USA
| | - Lisa D Coles
- University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Wenzhu B Mowrey
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Qianyun Li
- Albert Einstein College of Medicine, Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Bronx, NY, USA
| | - Wei Liu
- Albert Einstein College of Medicine, Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Bronx, NY, USA
| | - Solomon L Moshé
- Albert Einstein College of Medicine, Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Bronx, NY, USA; Isabelle Rapin Division of Child Neurology, Albert Einstein College of Medicine, Bronx, NY, USA; Dominick P Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Aristea S Galanopoulou
- Albert Einstein College of Medicine, Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Bronx, NY, USA; Isabelle Rapin Division of Child Neurology, Albert Einstein College of Medicine, Bronx, NY, USA; Dominick P Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
4
|
Chavda VP, Balar PC, Vaghela DA, Dodiya P. Unlocking longevity with GLP-1: A key to turn back the clock? Maturitas 2024; 186:108028. [PMID: 38815535 DOI: 10.1016/j.maturitas.2024.108028] [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: 01/17/2024] [Revised: 05/05/2024] [Accepted: 05/11/2024] [Indexed: 06/01/2024]
Abstract
Traditionally known for managing blood sugar, GLP-1, a gut hormone, is emerging as a potential key to both lengthening lifespan and combating age-related ailments. While widely recognized for its role in blood sugar control, GLP-1 is increasingly recognized for its diverse effects on various biological pathways beyond glucose metabolism. Research across organisms and humans suggests that activating GLP-1 receptors significantly impacts cellular processes linked to aging. Its ability to boost mitochondrial function, enhance cellular stress resistance, and quell inflammation hints at its wider influence on aging mechanisms. This intricate interplay between GLP-1 and longevity appears to act through multiple pathways. One key effect is its ability to modulate insulin sensitivity, potentially curbing age-related metabolic issues like type 2 diabetes. Its neuroprotective properties also make it a promising candidate for addressing age-related cognitive decline and neurodegenerative diseases. Furthermore, preclinical studies using GLP-1 analogs or agonists have shown promising results in extending lifespan and improving healthspan in various model organisms. These findings provide a compelling rationale for exploring GLP-1-based interventions in humans to extend healthy aging. However, despite the exciting therapeutic prospects of GLP-1 in promoting longevity, challenges remain. Determining optimal dosages, establishing long-term safety profiles, and investigating potential adverse effects require comprehensive clinical investigations before we can confidently translate these findings to humans. This article emphasises the wide applicability of GLP-1.
Collapse
Affiliation(s)
- Vivek P Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L M College of Pharmacy, Ahmedabad 380008, Gujarat, India.
| | - Pankti C Balar
- Pharmacy Section, L M College of Pharmacy, Ahmedabad 380008, Gujarat, India
| | - Dixa A Vaghela
- Pharmacy Section, L M College of Pharmacy, Ahmedabad 380008, Gujarat, India
| | - Payal Dodiya
- Pharmacy Section, L M College of Pharmacy, Ahmedabad 380008, Gujarat, India
| |
Collapse
|
5
|
Alharbi SH. Anti-inflammatory role of glucagon-like peptide 1 receptor agonists and its clinical implications. Ther Adv Endocrinol Metab 2024; 15:20420188231222367. [PMID: 38288136 PMCID: PMC10823863 DOI: 10.1177/20420188231222367] [Citation(s) in RCA: 54] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 11/28/2023] [Indexed: 01/31/2024] Open
Abstract
Glucagon-like peptide 1 receptor agonists (GLP-1RAs) have emerged as promising therapeutic agents with potent anti-inflammatory properties and diverse clinical implications. This in-depth review article explores the mechanisms behind the anti-inflammatory actions of GLP-1RAs and assesses their prospective applicability in a wide range of disease scenarios. The current review establishes the significance of comprehending the anti-inflammatory role of GLP-1RAs and identifies pertinent research gaps. A concise overview of inflammation and its clinical consequences underscores the critical need for effective anti-inflammatory interventions. Subsequently, the article elucidates the intricate mechanisms through which GLP-1RAs modulate immune cell signaling and regulate the nuclear factor-kappa B (NF-κB) pathway. Detailed discussions encompass their impact on inflammatory responses, cytokine production, and attenuation of oxidative stress. The exposition is substantiated by a collection of pertinent examples and an extensive array of references from both preclinical and clinical investigations. The historical trajectory of GLP-1RA drugs, including exenatide, lixisenatide, liraglutide, and semaglutide, is traced to delineate their development as therapeutic agents. Moreover, the review emphasizes the therapeutic potential of GLP-1RAs in specific disease contexts like type 2 diabetes, a neurodegenerative disorder, and inflammatory bowel disease (IBD), shedding light on their anti-inflammatory effects through rigorous examination of preclinical and clinical studies. The article also provides an outlook on future perspectives for GLP-1RAs, encompassing the domains of diabetes, neurodegenerative diseases, and IBD. In conclusion, GLP-1RAs exhibit substantial anti-inflammatory effects, rendering them promising therapeutic agents with broad clinical implications. They are very useful in a wide variety of diseases because they regulate immunological responses, block NF-κB activation, and decrease production of pro-inflammatory cytokines. Ongoing research endeavors aim to optimize their therapeutic use, delineate patient-specific treatment paradigms, and explore novel therapeutic applications. GLP-1RAs represent a significant breakthrough in anti-inflammatory therapy, offering novel treatment options, and improved patient outcomes.
Collapse
Affiliation(s)
- Saleh Hadi Alharbi
- Department of Medicine, Imam Mohammed Ibn Saud Islamic University, Riyadh 11652, Saudi Arabia
| |
Collapse
|
6
|
Sun W, Wang M, Zhao J, Zhao S, Zhu W, Wu X, Li F, Liu W, Wang Z, Gao M, Zhang Y, Xu J, Zhang M, Wang Q, Wen Z, Shen J, Zhang W, Huang Z. Sulindac selectively induces autophagic apoptosis of GABAergic neurons and alters motor behaviour in zebrafish. Nat Commun 2023; 14:5351. [PMID: 37660128 PMCID: PMC10475106 DOI: 10.1038/s41467-023-41114-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 08/22/2023] [Indexed: 09/04/2023] Open
Abstract
Nonsteroidal anti-inflammatory drugs compose one of the most widely used classes of medications, but the risks for early development remain controversial, especially in the nervous system. Here, we utilized zebrafish larvae to assess the potentially toxic effects of nonsteroidal anti-inflammatory drugs and found that sulindac can selectively induce apoptosis of GABAergic neurons in the brains of zebrafish larvae brains. Zebrafish larvae exhibit hyperactive behaviour after sulindac exposure. We also found that akt1 is selectively expressed in GABAergic neurons and that SC97 (an Akt1 activator) and exogenous akt1 mRNA can reverse the apoptosis caused by sulindac. Further studies showed that sulindac binds to retinoid X receptor alpha (RXRα) and induces autophagy in GABAergic neurons, leading to activation of the mitochondrial apoptotic pathway. Finally, we verified that sulindac can lead to hyperactivity and selectively induce GABAergic neuron apoptosis in mice. These findings suggest that excessive use of sulindac may lead to early neurodevelopmental toxicity and increase the risk of hyperactivity, which could be associated with damage to GABAergic neurons.
Collapse
Affiliation(s)
- Wenwei Sun
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Meimei Wang
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Jun Zhao
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Shuang Zhao
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Wenchao Zhu
- National Engineering Research Center for Tissue Restoration and Reconstruction, Key Laboratory of Biomedical Engineering of Guangdong Province, Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, Innovation Center for Tissue Restoration Reconstruction, South China University of Technology, Guangzhou, 510006, China
| | - Xiaoting Wu
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Feifei Li
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Wei Liu
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Zhuo Wang
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Meng Gao
- National Engineering Research Center for Tissue Restoration and Reconstruction, Key Laboratory of Biomedical Engineering of Guangdong Province, Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, Innovation Center for Tissue Restoration Reconstruction, South China University of Technology, Guangzhou, 510006, China
| | - Yiyue Zhang
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Jin Xu
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Meijia Zhang
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Qiang Wang
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Zilong Wen
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Center of Systems Biology and Human Health, the Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, People's Republic of China
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, 518055, China
| | - Juan Shen
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| | - Wenqing Zhang
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, 510006, China.
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen, 518055, China.
| | - Zhibin Huang
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, 510006, China.
| |
Collapse
|
7
|
Gao M, Wang K, Zhao H. GABAergic neurons maturation is regulated by a delicate network. Int J Dev Neurosci 2023; 83:3-15. [PMID: 36401305 DOI: 10.1002/jdn.10242] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 10/25/2022] [Accepted: 11/13/2022] [Indexed: 11/21/2022] Open
Abstract
Gamma-aminobutyric acid-expressing (GABAergic) neurons are implicated in a variety of neuropsychiatric disorders, such as epilepsy, anxiety, autism, and other pathological processes, including cerebral ischemia injury and drug addiction. Therefore, GABAergic neuronal processes warrant further research. The development of GABAergic neurons is a tightly controlled process involving the activity of multiple transcription and growth factors. Here, we focus on the gene expression pathways and the molecular modulatory networks that are engaged during the development of GABAergic neurons with the goal of exploring regulatory mechanisms that influence GABAergic neuron fate (i.e., maturation). Overall, we hope to provide a basis for clarifying the pathogenesis of neurodegenerative disorders.
Collapse
Affiliation(s)
- Mingxing Gao
- Department of Histology and Embryology, School of Basic Medical Science, Jilin University, Changchun, Jilin, China
| | - Kaizhong Wang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Hui Zhao
- Department of Histology and Embryology, School of Basic Medical Science, Jilin University, Changchun, Jilin, China
| |
Collapse
|
8
|
Diaz C, de la Torre MM, Rubenstein JLR, Puelles L. Dorsoventral Arrangement of Lateral Hypothalamus Populations in the Mouse Hypothalamus: a Prosomeric Genoarchitectonic Analysis. Mol Neurobiol 2023; 60:687-731. [PMID: 36357614 PMCID: PMC9849321 DOI: 10.1007/s12035-022-03043-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 09/16/2022] [Indexed: 11/12/2022]
Abstract
The lateral hypothalamus (LH) has a heterogeneous cytoarchitectonic organization that has not been elucidated in detail. In this work, we analyzed within the framework of the prosomeric model the differential expression pattern of 59 molecular markers along the ventrodorsal dimension of the medial forebrain bundle in the mouse, considering basal and alar plate subregions of the LH. We found five basal (LH1-LH5) and four alar (LH6-LH9) molecularly distinct sectors of the LH with neuronal cell groups that correlate in topography with previously postulated alar and basal hypothalamic progenitor domains. Most peptidergic populations were restricted to one of these LH sectors though some may have dispersed into a neighboring sector. For instance, histaminergic Hdc-positive neurons were mostly contained within the basal LH3, Nts (neurotensin)- and Tac2 (tachykinin 2)-expressing cells lie strictly within LH4, Hcrt (hypocretin/orexin)-positive and Pmch (pro-melanin-concentrating hormone)-positive neurons appeared within separate LH5 subdivisions, Pnoc (prepronociceptin)-expressing cells were mainly restricted to LH6, and Sst (somatostatin)-positive cells were identified within the LH7 sector. The alar LH9 sector, a component of the Foxg1-positive telencephalo-opto-hypothalamic border region, selectively contained Satb2-expressing cells. Published studies of rodent LH subdivisions have not described the observed pattern. Our genoarchitectonic map should aid in systematic approaches to elucidate LH connectivity and function.
Collapse
Affiliation(s)
- Carmen Diaz
- Department of Medical Sciences, School of Medicine and Institute for Research in Neurological Disabilities, University of Castilla-La Mancha, 02006 Albacete, Spain
| | - Margaret Martinez de la Torre
- Department of Human Anatomy and Psychobiology and IMIB-Arrixaca Institute, University of Murcia, 30100 Murcia, Spain
| | - John L. R. Rubenstein
- Nina Ireland Laboratory of Developmental Neurobiology, Department of Psychiatry, UCSF Medical School, San Francisco, California USA
| | - Luis Puelles
- Department of Human Anatomy and Psychobiology and IMIB-Arrixaca Institute, University of Murcia, 30100 Murcia, Spain
| |
Collapse
|
9
|
Cheng D, Yang S, Zhao X, Wang G. The Role of Glucagon-Like Peptide-1 Receptor Agonists (GLP-1 RA) in Diabetes-Related Neurodegenerative Diseases. Drug Des Devel Ther 2022; 16:665-684. [PMID: 35340338 PMCID: PMC8943601 DOI: 10.2147/dddt.s348055] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 02/18/2022] [Indexed: 12/17/2022] Open
Abstract
Recent clinical guidelines have emphasized the importance of screening for cognitive impairment in older adults with diabetes, however, there is still a lack of understanding about the drug therapy. Glucagon-like peptide 1 receptor agonists (GLP-1 RAs) are widely used in the treatment of type 2 diabetes and potential applications may include the treatment of obesity as well as the adjunctive treatment of type 1 diabetes mellitus in combination with insulin. Growing evidence suggests that GLP-1 RA has the potential to treat neurodegenerative diseases, particularly in diabetes-related Alzheimer’s disease (AD) and Parkinson’s disease (PD). Here, we review the molecular mechanisms of the neuroprotective effects of GLP-1 RA in diabetes-related degenerative diseases, including AD and PD, and their potential effects.
Collapse
Affiliation(s)
- Dihe Cheng
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, People's Republic of China
| | - Shuo Yang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, People's Republic of China
| | - Xue Zhao
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, People's Republic of China
| | - Guixia Wang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, People's Republic of China
| |
Collapse
|
10
|
Han X, Wei Y, Ba R, Sun L, Zhao C. PDK1 Regulates the Lengthening of G1 Phase to Balance RGC Proliferation and Differentiation during Cortical Neurogenesis. Cereb Cortex 2021; 32:3488-3500. [PMID: 34918060 DOI: 10.1093/cercor/bhab428] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 11/02/2021] [Accepted: 11/02/2021] [Indexed: 12/11/2022] Open
Abstract
During cortical development, the balance between progenitor self-renewal and neurogenesis is critical for determining the size/morphology of the cortex. A fundamental feature of the developing cortex is an increase in the length of G1 phase in RGCs over the course of neurogenesis, which is a key determinant of progenitor fate choice. How the G1 length is temporally regulated remains unclear. Here, Pdk1, a member of the AGC kinase family, was conditionally disrupted by crossing an Emx1-Cre mouse line with a Pdk1fl/fl line. The loss of Pdk1 led to a shorter cell cycle accompanied by increased RGC proliferation specifically at late rather than early/middle neurogenic stages, which was attributed to impaired lengthening of G1 phase. Coincidently, apical-to-basal interkinetic nuclear migration was accelerated in Pdk1 cKO cortices. Consequently, we detected an increased neuronal output at P0. We further showed the significant upregulation of the cell cycle regulator cyclin D1 and its activator Myc in the cKO cortices relative to those of control animals. Overall, we have identified a novel role for PDK1 in cortical neurogenesis. PDK1 functions as an upstream regulator of the Myc-cyclin D1 pathway to control the lengthening of G1 phase and the balance between RGC proliferation and differentiation.
Collapse
Affiliation(s)
- Xiaoning Han
- Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, School of Medicine, Southeast University, Nanjing 210009, China.,Institute of Biomedical Engineering and Health Science, Changzhou University, Changzhou 213164, China
| | - Yongjie Wei
- Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, School of Medicine, Southeast University, Nanjing 210009, China
| | - Ru Ba
- Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, School of Medicine, Southeast University, Nanjing 210009, China
| | - Lijuan Sun
- Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, School of Medicine, Southeast University, Nanjing 210009, China
| | - Chunjie Zhao
- Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, School of Medicine, Southeast University, Nanjing 210009, China
| |
Collapse
|
11
|
Rana T, Behl T, Sehgal A, Mehta V, Singh S, Sharma N, Bungau S. Elucidating the Possible Role of FoxO in Depression. Neurochem Res 2021; 46:2761-2775. [PMID: 34075521 DOI: 10.1007/s11064-021-03364-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/23/2021] [Accepted: 05/25/2021] [Indexed: 12/21/2022]
Abstract
Forkhead box-O (FoxO) transcriptional factors perform essential functions in several physiological and biological processes. Recent studies have shown that FoxO is implicated in the pathophysiology of depression. Changes in the upstream mediators of FoxOs including brain-derived neurotrophic factor (BDNF) and protein kinase B have been associated with depressive disorder and the antidepressant agents are known to alter the phosphorylation of FoxOs. Moreover, FoxOs might be regulated by serotonin or noradrenaline signaling and the hypothalamic-pituitary-adrenal (HPA)-axis,both of them are associated with the development of the depressive disorder. FoxO also regulates neural morphology, synaptogenesis, and neurogenesis in the hippocampus, which accounts for the pathogenesis of the depressive disorder. The current article underlined the potential functions of FoxOs in the etiology of depressive disorder and formulate few essential proposals for further investigation. The review also proposes that FoxO and its signal pathway might establish possible therapeutic mediators for the management of depressive disorder.
Collapse
Affiliation(s)
- Tarapati Rana
- Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India.,Government Pharmacy College, Seraj, Mandi, Himachal Pradesh, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India.
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India
| | - Vineet Mehta
- Government College of Pharmacy, Rohru, Distt., Shimla, Himachal Pradesh, India
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India
| | - Neelam Sharma
- Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
12
|
Gene Profiles in the Early Stage of Neuronal Differentiation of Mouse Bone Marrow Stromal Cells Induced by Basic Fibroblast Growth Factor. Stem Cells Int 2021; 2020:8857057. [PMID: 33424980 PMCID: PMC7775150 DOI: 10.1155/2020/8857057] [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: 04/28/2020] [Revised: 11/30/2020] [Accepted: 12/08/2020] [Indexed: 11/20/2022] Open
Abstract
A stably established population of mouse bone marrow stromal cells (BMSCs) with self-renewal and multilineage differentiation potential was expanded in vitro for more than 50 passages. These cells express high levels of mesenchymal stem cell markers and can be differentiated into adipogenic, chondrogenic, and osteogenic lineages in vitro. Subjected to basic fibroblast growth factor (bFGF) treatment, a typical neuronal phenotype was induced in these cells, as supported by neuronal morphology, induction of neuronal markers, and relevant electrophysiological excitability. To identify the genes regulating neuronal differentiation, cDNA microarray analysis was conducted using mRNAs isolated from cells differentiated for different time periods (0, 4, 24, and 72 h) after bFGF treatment. Various expression patterns of neuronal genes were stimulated by bFGF. These gene profiles were shown to be involved in developmental, functional, and structural integration of the nervous system. The expression of representative genes stimulated by bFGF in each group was verified by RT-PCR. Amongst proneural genes, the mammalian achate-schute homolog 1 (Mash-1), a basic helix-loop-helix transcriptional factor, was further demonstrated to be significantly upregulated. Overexpression of Mash-1 in mouse BMSCs was shown to induce the expression of neuronal specific enolase (NSE) and terminal neuronal morphology, suggesting that Mash-1 plays an important role in the induction of neuronal differentiation of mouse BMSCs.
Collapse
|
13
|
Lim SH, Shin S, Kim MH, Kim EC, Lee DY, Moon J, Park HY, Ryu YK, Kang YM, Kang YJ, Kim TH, Lee NY, Kim NS, Yu DY, Shim I, Gondo Y, Satake M, Kim E, Kim KS, Min SS, Lee JR. Depression-like behaviors induced by defective PTPRT activity through dysregulated synaptic functions and neurogenesis. J Cell Sci 2020; 133:jcs243972. [PMID: 32938684 DOI: 10.1242/jcs.243972] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 09/07/2020] [Indexed: 12/27/2022] Open
Abstract
PTPRT has been known to regulate synaptic formation and dendritic arborization of hippocampal neurons. PTPRT-/- null and PTPRT-D401A mutant mice displayed enhanced depression-like behaviors compared with wild-type mice. Transient knockdown of PTPRT in the dentate gyrus enhanced the depression-like behaviors of wild-type mice, whereas rescued expression of PTPRT ameliorated the behaviors of PTPRT-null mice. Chronic stress exposure reduced expression of PTPRT in the hippocampus of mice. In PTPRT-deficient mice the expression of GluR2 (also known as GRIA2) was attenuated as a consequence of dysregulated tyrosine phosphorylation, and the long-term potentiation at perforant-dentate gyrus synapses was augmented. The inhibitory synaptic transmission of the dentate gyrus and hippocampal GABA concentration were reduced in PTPRT-deficient mice. In addition, the hippocampal expression of GABA transporter GAT3 (also known as SLC6A11) was decreased, and its tyrosine phosphorylation was increased in PTPRT-deficient mice. PTPRT-deficient mice displayed reduced numbers and neurite length of newborn granule cells in the dentate gyrus and had attenuated neurogenic ability of embryonic hippocampal neural stem cells. In conclusion, our findings show that the physiological roles of PTPRT in hippocampal neurogenesis, as well as synaptic functions, are involved in the pathogenesis of depressive disorder.
Collapse
Affiliation(s)
- So-Hee Lim
- Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
- Department of Biological Sciences, Chungnam National University, Daejeon 34134, Korea
| | - Sangyep Shin
- Department of Physiology and Biophysics, School of Medicine, Eulji University, Daejeon 34824, Korea
| | - Myoung-Hwan Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Eung Chang Kim
- Department of Physiology and Biophysics, School of Medicine, Eulji University, Daejeon 34824, Korea
| | - Da Yong Lee
- Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
| | - Jeonghee Moon
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
| | - Hye-Yeon Park
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
| | - Young-Kyoung Ryu
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
| | - Young-Mi Kang
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
| | - Yu Jeong Kang
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
| | - Tae Hwan Kim
- Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
| | - Na-Yoon Lee
- Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
| | - Nam-Soon Kim
- Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
| | - Dae-Yeul Yu
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
| | - Insop Shim
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Yoichi Gondo
- Department of Molecular Life Sciences, Tokai University School of Medicine, Shimo-Kasuya, Isehara 259-1193, Japan
| | - Masanobu Satake
- Department of Molecular Immunology, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | - Eunhee Kim
- Department of Biological Sciences, Chungnam National University, Daejeon 34134, Korea
| | - Kyoung-Shim Kim
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
| | - Sun Seek Min
- Department of Physiology and Biophysics, School of Medicine, Eulji University, Daejeon 34824, Korea
| | - Jae-Ran Lee
- Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
| |
Collapse
|
14
|
PDK1 Regulates the Maintenance of Cell Body and the Development of Dendrites of Purkinje Cells by pS6 and PKCγ. J Neurosci 2020; 40:5531-5548. [PMID: 32487697 DOI: 10.1523/jneurosci.2496-19.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 01/09/2023] Open
Abstract
3-Phosphoinositide-dependent protein kinase-1 (PDK1) plays a critical role in the development of mammalian brain. Here, we investigated the role of PDK1 in Purkinje cells (PCs) by generating the PDK1-conditional knock-out mice (cKO) through crossing PV-cre or Pcp2-cre mice with Pdk1fl/fl mice. The male mice were used in the behavioral testing, and the other experiments were performed on mice of both sexes. These PDK1-cKO mice displayed decreased cerebellar size and impaired motor balance and coordination. By the electrophysiological recording, we observed the reduced spontaneous firing of PCs from the cerebellar slices of the PDK1-cKO mice. Moreover, the cell body size of PCs in the PDK1-cKO mice was time dependently reduced compared with that in the control mice. And the morphologic complexity of PCs was also decreased after PDK1 deletion. These effects may have contributed to the reduction of the rpS6 (reduced ribosomal protein S6) phosphorylation and the PKCγ expression in PDK1-cKO mice since the upregulation of pS6 by treatment of 3-benzyl-5-((2-nitrophenoxy) methyl)-dihydrofuran-2(3H)-1, the agonist of mTOR1, partly rescued the reduction in the cell body size of the PCs, and the delivery of recombinant adeno-associated virus-PKCγ through cerebellar injection rescued the reduced complexity of the dendritic arbor in PDK1-cKO mice. Together, our data suggest that PDK1, by regulating rpS6 phosphorylation and PKCγ expression, controls the cell body maintenance and the dendritic development in PCs and is critical for cerebellar motor coordination.SIGNIFICANCE STATEMENT Here, we show the role of 3-phosphoinositide-dependent protein kinase-1 (PDK1) in Purkinje cells (PCs). The ablation of PDK1 in PCs resulted in a reduction of cell body size, and dendritic complexity and abnormal spontaneous firing, which attributes to the motor defects in PDK1-conditional knock-out (cKO) mice. Moreover, the ribosomal protein S6 (rpS6) phosphorylation and the expression of PKCγ are downregulated after the ablation of PDK1. Additionally, upregulation of rpS6 phosphorylation by3-benzyl-5-((2-nitrophenoxy) methyl)-dihydrofuran-2(3H)-1 partly rescued the reduction in cell body size of PCs, and the overexpression of PKCγ in PDK1-KO PCs rescued the reduction in the dendritic complexity. These findings indicate that PDK1 contributes to the maintenance of the cell body and the dendritic development of PCs by regulating rpS6 phosphorylation and PKCγ expression.
Collapse
|
15
|
Wei Y, Han X, Zhao C. PDK1 regulates the survival of the developing cortical interneurons. Mol Brain 2020; 13:65. [PMID: 32366272 PMCID: PMC7197138 DOI: 10.1186/s13041-020-00604-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 04/22/2020] [Indexed: 01/08/2023] Open
Abstract
Inhibitory interneurons are critical for maintaining the excitatory/inhibitory balance. During the development cortical interneurons originate from the ganglionic eminence and arrive at the dorsal cortex through two tangential migration routes. However, the mechanisms underlying the development of cortical interneurons remain unclear. 3-Phosphoinositide-dependent protein kinase-1 (PDK1) has been shown to be involved in a variety of biological processes, including cell proliferation and migration, and plays an important role in the neurogenesis of cortical excitatory neurons. However, the function of PDK1 in interneurons is still unclear. Here, we reported that the disruption of Pdk1 in the subpallium achieved by crossing the Dlx5/6-Cre-IRES-EGFP line with Pdk1fl/fl mice led to the severely increased apoptosis of immature interneurons, subsequently resulting in a remarkable reduction in cortical interneurons. However, the tangential migration, progenitor pools and cell proliferation were not affected by the disruption of Pdk1. We further found the activity of AKT-GSK3β signaling pathway was decreased after Pdk1 deletion, suggesting it might be involved in the regulation of the survival of cortical interneurons. These results provide new insights into the function of PDK1 in the development of the telencephalon.
Collapse
Affiliation(s)
- Yongjie Wei
- Key Laboratory of Developmental Genes and Human Diseases, MOE, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Xiaoning Han
- Key Laboratory of Developmental Genes and Human Diseases, MOE, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Chunjie Zhao
- Key Laboratory of Developmental Genes and Human Diseases, MOE, School of Medicine, Southeast University, Nanjing, 210009, China.
| |
Collapse
|
16
|
Castilla-Cortázar I, Aguirre GA, Femat-Roldán G, Martín-Estal I, Espinosa L. Is insulin-like growth factor-1 involved in Parkinson's disease development? J Transl Med 2020; 18:70. [PMID: 32046737 PMCID: PMC7014772 DOI: 10.1186/s12967-020-02223-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 01/10/2020] [Indexed: 02/09/2023] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder that results in the death of dopaminergic neurons within the substantia nigra pars compacta and the reduction in dopaminergic control over striatal output neurons, leading to a movement disorder most commonly characterized by akinesia or bradykinesia, rigidity and tremor. Also, PD is less frequently depicted by sensory symptoms (pain and tingling), hyposmia, sleep alterations, depression and anxiety, and abnormal executive and working memory related functions. On the other hand, insulin-like growth factor 1 (IGF-1) is an endocrine, paracrine and autocrine hormone with several functions including tissue growth and development, insulin-like activity, proliferation, pro-survival, anti-aging, antioxidant and neuroprotection, among others. Herein this review tries to summarize all experimental and clinical data to understand the pathophysiology and development of PD, as well as its clear association with IGF-1, supported by several lines of evidence: (1) IGF-1 decreases with age, while aging is the major risk for PD establishment and development; (2) numerous basic and translational data have appointed direct protective and homeostasis IGF-1 roles in all brain cells; (3) estrogens seem to confer women strong protection to PD via IGF-1; and (4) clinical correlations in PD cohorts have confirmed elevated IGF-1 levels at the onset of the disease, suggesting an ongoing compensatory or "fight-to-injury" mechanism.
Collapse
Affiliation(s)
- Inma Castilla-Cortázar
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, 64710, Monterrey, N.L., Mexico.
- Fundación de Investigación HM Hospitales, Madrid, Spain.
| | - Gabriel A Aguirre
- Centre for Tumour Biology, Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Giovana Femat-Roldán
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, 64710, Monterrey, N.L., Mexico
- Neurocenter, Monterrey, Nuevo Leon, Mexico
| | - Irene Martín-Estal
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, 64710, Monterrey, N.L., Mexico
| | - Luis Espinosa
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, 64710, Monterrey, N.L., Mexico
| |
Collapse
|
17
|
Lithium for schizophrenia: supporting evidence from a 12-year, nationwide health insurance database and from Akt1-deficient mouse and cellular models. Sci Rep 2020; 10:647. [PMID: 31959776 PMCID: PMC6971245 DOI: 10.1038/s41598-019-57340-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 12/19/2019] [Indexed: 12/11/2022] Open
Abstract
Accumulating evidence suggests AKT1 and DRD2-AKT-GSK3 signaling involvement in schizophrenia. AKT1 activity is also required for lithium, a GSK3 inhibitor, to modulate mood-related behaviors. Notably, GSK3 inhibitor significantly alleviates behavioral deficits in Akt1−/− female mice, whereas typical/atypical antipsychotics have no effect. In agreement with adjunctive therapy with lithium in treating schizophrenia, our data mining indicated that the average utilization rates of lithium in the Taiwan National Health Insurance Research Database from 2002 to 2013 are 10.9% and 6.63% in inpatients and outpatients with schizophrenia, respectively. Given that lithium is commonly used in clinical practice, it is of great interest to evaluate the effect of lithium on alleviating Akt1-related deficits. Taking advantage of Akt1+/− mice to mimic genetic deficiency in patients, behavioral impairments were replicated in female Akt1+/− mice but were alleviated by subchronic lithium treatment for 13 days. Lithium also effectively alleviated the observed reduction in phosphorylated GSK3α/β expression in the brains of Akt1+/− mice. Furthermore, inhibition of Akt expression using an Akt1/2 inhibitor significantly reduced neurite length in P19 cells and primary hippocampal cell cultures, which was also ameliorated by lithium. Collectively, our findings implied the therapeutic potential of lithium and the importance of the AKT1-GSK3 signaling pathway.
Collapse
|
18
|
Han X, Wei Y, Wu X, Gao J, Yang Z, Zhao C. PDK1 Regulates Transition Period of Apical Progenitors to Basal Progenitors by Controlling Asymmetric Cell Division. Cereb Cortex 2019; 30:406-420. [DOI: 10.1093/cercor/bhz146] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/09/2019] [Accepted: 06/09/2019] [Indexed: 12/18/2022] Open
Abstract
Abstract
The six-layered neocortex consists of diverse neuron subtypes. Deeper-layer neurons originate from apical progenitors (APs), while upper-layer neurons are mainly produced by basal progenitors (BPs), which are derivatives of APs. As development proceeds, an AP generates two daughter cells that comprise an AP and a deeper-layer neuron or a BP. How the transition of APs to BPs is spatiotemporally regulated is a fundamental question. Here, we report that conditional deletion of phoshpoinositide-dependent protein kinase 1 (PDK1) in mouse developing cortex achieved by crossing Emx1Cre line with Pdk1fl/fl leads to a delayed transition of APs to BPs and subsequently causes an increased output of deeper-layer neurons. We demonstrate that PDK1 is involved in the modulation of the aPKC-Par3 complex and further regulates the asymmetric cell division (ACD). We also find Hes1, a downstream effecter of Notch signal pathway is obviously upregulated. Knockdown of Hes1 or treatment with Notch signal inhibitor DAPT recovers the ACD defect in the Pdk1 cKO. Thus, we have identified a novel function of PDK1 in controlling the transition of APs to BPs.
Collapse
Affiliation(s)
- Xiaoning Han
- Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, School of Medicine, Southeast University, Nanjing 210009, China
| | - Yongjie Wei
- Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, School of Medicine, Southeast University, Nanjing 210009, China
| | - Xiaojing Wu
- Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, School of Medicine, Southeast University, Nanjing 210009, China
| | - Jun Gao
- Department of Neurobiology
- Key Laboratory of Human Functional Genomics of Jiangsu, Nanjing Medical University, Nanjing 211166, China
| | - Zhongzhou Yang
- State Key Laboratory of Pharmaceutical Biotechnology
- Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing 210061, China
| | - Chunjie Zhao
- Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, School of Medicine, Southeast University, Nanjing 210009, China
| |
Collapse
|
19
|
Dong ZY, Pei Z, Wang YL, Li Z, Khan A, Meng XT. Ascl1 Regulates Electric Field-Induced Neuronal Differentiation Through PI3K/Akt Pathway. Neuroscience 2019; 404:141-152. [PMID: 30771509 DOI: 10.1016/j.neuroscience.2019.02.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 01/31/2019] [Accepted: 02/04/2019] [Indexed: 12/14/2022]
Abstract
Directing differentiation of neural stem/progenitor cells (NSCs/NPCs) to produce functional neurons is one of the greatest challenges in regenerative medicine. Our previous paper has confirmed that electrical stimulation has a high efficiency of triggering neuronal differentiation by using isolated filum terminale (FT)-derived NPCs. To further clarify the intrinsic molecular mechanisms, protein-protein interaction (PPI) network analysis was applied to pinpoints novel hubs in electric field (EF)-induced neuronal differentiation. In this study, siRNA transfection of Achaete-scute homolog 1 (Ascl1) in NPCs or NPCs was followed by direct current stimulation at 150 mV/mm. Neuronal differentiation rate and protein expression level were analyzed after 7 or 14 days of electrical stimulation. The data showed that the expression level of Ascl1 was enhanced by electrical stimulation and positively correlated to EF strength. Moreover, we identified that the expression of Ascl1 positively regulated neuronal differentiation of NPCs and can be up-regulated by EF-stimulation through the activation of phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) pathway. Therefore, this study provides new insights into the role of Ascl1 and its relevant PI3K/Akt pathway in regulating of EF-induced neuronal differentiation and pointed out that continuous expression of Ascl1 in NPCs is required for EF-induced neuronal differentiation.
Collapse
Affiliation(s)
- Zhi-Yong Dong
- Department of Histology & Embryology, College of Basic Medical Sciences, Jilin University, Changchun 130021, PR China.
| | - Zhe Pei
- Department of Neuroscience and Pediatric, GSRB1 Duke University, Durham 27710, USA
| | - Yan-Ling Wang
- Laboratory Teaching Center of Basic Medicine, College of Basic Medical Sciences, Jilin University, Changchun 130021, PR China.
| | - Zhe Li
- Laboratory Teaching Center of Basic Medicine, College of Basic Medical Sciences, Jilin University, Changchun 130021, PR China.
| | - Amber Khan
- The Graduate Center and CUNY School of Medicine, CUNY, 85 St Nicholas Terrace, New York, NY 10027, USA.
| | - Xiao-Ting Meng
- Department of Histology & Embryology, College of Basic Medical Sciences, Jilin University, Changchun 130021, PR China.
| |
Collapse
|
20
|
Ishimoto T, Masuo Y, Kato Y, Nakamichi N. Ergothioneine-induced neuronal differentiation is mediated through activation of S6K1 and neurotrophin 4/5-TrkB signaling in murine neural stem cells. Cell Signal 2018; 53:269-280. [PMID: 30359715 DOI: 10.1016/j.cellsig.2018.10.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 10/15/2018] [Accepted: 10/18/2018] [Indexed: 01/08/2023]
Abstract
The promotion of neurogenesis is considered to be an effective therapeutic strategy for neuropsychiatric disorders because impairment of neurogenesis is associated with the onset and progression of these disorders. We have previously demonstrated that orally ingested ergothioneine (ERGO), a naturally occurring antioxidant and hydrophilic amino acid, promotes neurogenesis in the hippocampal dentate gyrus (DG) with its abundant neural stem cells (NSCs) and exerts antidepressant-like effects in mice. Independent of its antioxidant activities, ERGO induces in cultured NSCs this differentiation through induction of the basic helix-loop-helix transcription factor Math1. However, the upstream signaling of Math1 in the mechanisms underlying ERGO-induced neuronal differentiation remains unclear. The purpose of the present study was to elucidate the upstream signaling with the aim of discovering novel targets for the treatment of neuropsychiatric disorders. We focused on neurotrophic factor signaling, as it is important for the promotion of neurogenesis and the induction of antidepressant effects. We also focused on the signaling of the mammalian target of rapamycin (mTOR) complex 1 (mTORC1), a known amino acid sensor, and the members of this signaling pathway, mTOR and p70 ribosomal protein S6 kinase 1 (S6K1). Exposure of cultured NSCs to ERGO significantly increased the expression of phosphorylated S6K1 (p-S6K1) at Thr389 in only 1 h, of phosphorylated mTOR (p-mTOR) in 6 h, and of the gene product of neurotrophin 4/5 (NT5) which activates tropomyosin receptor kinase B (TrkB) in 24 h. ERGO increased the population of βIII-tubulin-positive neurons, and this effect was suppressed by the inhibitors of S6K1 (PF4708671), mTORC1 (rapamycin), and TrkB (GNF5837). Oral administration of ERGO to mice significantly increased in the DG the expression of p-S6K1 at Thr389, the gene product of NT5, and phosphorylated TrkB but not that of p-mTOR. Thus, neuronal differentiation of NSCs induced by ERGO is mediated, at least in part, through phosphorylation of S6K1 at Thr389 and subsequent activation of TrkB signaling through the induction of NT5. Thus, S6K1 and NT5 might be promising target molecules for the treatment of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Takahiro Ishimoto
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
| | - Yusuke Masuo
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
| | - Yukio Kato
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
| | - Noritaka Nakamichi
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan.
| |
Collapse
|
21
|
Lin C, Chen P, Chan H, Huang Y, Chang NW. Peroxisome proliferator‐activated receptor alpha accelerates neuronal differentiation and this might involve the mitogen‐activated protein kinase pathway. Int J Dev Neurosci 2018; 71:46-51. [DOI: 10.1016/j.ijdevneu.2018.08.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 08/09/2018] [Accepted: 08/21/2018] [Indexed: 01/11/2023] Open
Affiliation(s)
- Chingju Lin
- Department of PhysiologyCollege of Medicine, China Medical UniversityTaichungTaiwan, ROC
| | - Pei‐Yi Chen
- Department of BiochemistryCollege of Medicine, China Medical UniversityTaichungTaiwan, ROC
| | - Hsu‐Chin Chan
- Department of BiochemistryCollege of Medicine, China Medical UniversityTaichungTaiwan, ROC
| | - Yi‐Ping Huang
- Department of PhysiologyCollege of Medicine, China Medical UniversityTaichungTaiwan, ROC
| | - Nai Wen Chang
- Department of BiochemistryCollege of Medicine, China Medical UniversityTaichungTaiwan, ROC
| |
Collapse
|
22
|
Zucco AJ, Pozzo VD, Afinogenova A, Hart RP, Devinsky O, D'Arcangelo G. Neural progenitors derived from Tuberous Sclerosis Complex patients exhibit attenuated PI3K/AKT signaling and delayed neuronal differentiation. Mol Cell Neurosci 2018; 92:149-163. [PMID: 30144504 DOI: 10.1016/j.mcn.2018.08.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 07/23/2018] [Accepted: 08/20/2018] [Indexed: 12/20/2022] Open
Abstract
Tuberous Sclerosis Complex (TSC) is a disease caused by autosomal dominant mutations in the TSC1 or TSC2 genes, and is characterized by tumor susceptibility, brain lesions, seizures and behavioral impairments. The TSC1 and TSC2 genes encode proteins forming a complex (TSC), which is a major regulator and suppressor of mammalian target of rapamycin complex 1 (mTORC1), a signaling complex that promotes cell growth and proliferation. TSC1/2 loss of heterozygosity (LOH) and the subsequent complete loss of TSC regulatory activity in null cells causes mTORC1 dysregulation and TSC-associated brain lesions or other tissue tumors. However, it is not clear whether TSC1/2 heterozygous brain cells are abnormal and contribute to TSC neuropathology. To investigate this issue, we generated induced pluripotent stem cells (iPSCs) from TSC patients and unaffected controls, and utilized these to obtain neural progenitor cells (NPCs) and differentiated neurons in vitro. These patient-derived TSC2 heterozygous NPCs were delayed in their ability to differentiate into neurons. Patient-derived progenitor cells also exhibited a modest activation of mTORC1 signaling downstream of TSC, and a marked attenuation of upstream PI3K/AKT signaling. We further show that pharmacologic PI3K or AKT inhibition, but not mTORC1 inhibition, causes a neuronal differentiation delay, mimicking the patient phenotype. Together these data suggest that heterozygous TSC2 mutations disrupt neuronal development, potentially contributing to the disease neuropathology, and that this defect may result from dysregulated PI3K/AKT signaling in neural progenitor cells.
Collapse
Affiliation(s)
- Avery J Zucco
- Graduate Program in Neuroscience, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, United States of America; Department of Cell Biology and Neuroscience, Rutgers, the State University of New Jersey, Piscataway, NJ, United States of America
| | - Valentina Dal Pozzo
- Graduate Program in Neuroscience, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, United States of America; Department of Cell Biology and Neuroscience, Rutgers, the State University of New Jersey, Piscataway, NJ, United States of America
| | - Alina Afinogenova
- Department of Cell Biology and Neuroscience, Rutgers, the State University of New Jersey, Piscataway, NJ, United States of America
| | - Ronald P Hart
- Department of Cell Biology and Neuroscience, Rutgers, the State University of New Jersey, Piscataway, NJ, United States of America; Human Genetics Institute of New Jersey, Piscataway, NJ, United States of America
| | - Orrin Devinsky
- NYU Comprehensive Epilepsy Center, NYU Langone School of Medicine, New York, NY, United States of America
| | - Gabriella D'Arcangelo
- Department of Cell Biology and Neuroscience, Rutgers, the State University of New Jersey, Piscataway, NJ, United States of America; Human Genetics Institute of New Jersey, Piscataway, NJ, United States of America.
| |
Collapse
|
23
|
Xu M, Han X, Liu R, Li Y, Qi C, Yang Z, Zhao C, Gao J. PDK1 Deficit Impairs the Development of the Dentate Gyrus in Mice. Cereb Cortex 2018; 29:1185-1198. [DOI: 10.1093/cercor/bhy024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Indexed: 12/22/2022] Open
Affiliation(s)
- Min Xu
- Department of Neurobiology, Nanjing Medical University, Nanjing, China
- Key Laboratory of Human Functional Genomics of Jiangsu, Nanjing Medical University, Nanjing, China
| | - Xiaoning Han
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, China
| | - Rui Liu
- Department of Neurobiology, Nanjing Medical University, Nanjing, China
- Key Laboratory of Human Functional Genomics of Jiangsu, Nanjing Medical University, Nanjing, China
| | - Yanjun Li
- Model Animal Research Center of Nanjing University and MOE Key Laboratory of Model Animals for Disease Study, Nanjing, China
| | - Cui Qi
- Department of Neurobiology, Nanjing Medical University, Nanjing, China
- Key Laboratory of Human Functional Genomics of Jiangsu, Nanjing Medical University, Nanjing, China
| | - Zhongzhou Yang
- Model Animal Research Center of Nanjing University and MOE Key Laboratory of Model Animals for Disease Study, Nanjing, China
| | - Chunjie Zhao
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, China
- Center of Depression, Beijing Institute for Brain Disorders, Beijing, China
| | - Jun Gao
- Department of Neurobiology, Nanjing Medical University, Nanjing, China
- Key Laboratory of Human Functional Genomics of Jiangsu, Nanjing Medical University, Nanjing, China
| |
Collapse
|
24
|
Narducci R, Baroncelli L, Sansevero G, Begenisic T, Prontera C, Sale A, Cenni MC, Berardi N, Maffei L. Early impoverished environment delays the maturation of cerebral cortex. Sci Rep 2018; 8:1187. [PMID: 29352131 PMCID: PMC5775315 DOI: 10.1038/s41598-018-19459-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 12/27/2017] [Indexed: 12/26/2022] Open
Abstract
The influence of exposure to impoverished environments on brain development is unexplored since most studies investigated how environmental impoverishment affects adult brain. To shed light on the impact of early impoverishment on developmental trajectories of the nervous system, we developed a protocol of environmental impoverishment in which dams and pups lived from birth in a condition of reduced sensory-motor stimulation. Focusing on visual system, we measured two indexes of functional development, that is visual acuity, assessed by using Visual Evoked Potentials (VEPs), and VEP latency. In addition, we assessed in the visual cortex levels of Insulin-Like Growth Factor 1 (IGF-1) and myelin maturation, together with the expression of the GABA biosynthetic enzyme GAD67. We found that early impoverishment strongly delays visual acuity and VEP latency development. These functional changes were accompanied by a significant reduction of IGF-1 protein and GAD67 expression, as well as by delayed myelination of nerve fibers, in the visual cortex of impoverished pups. Thus, exposure to impoverished living conditions causes a significant alteration of developmental trajectories leading to a prominent delay of brain maturation. These results underscore the significance of adequate levels of environmental stimulation for the maturation of central nervous system.
Collapse
Affiliation(s)
- Roberta Narducci
- Institute of Neuroscience, National Research Council (CNR), Via Moruzzi 1, I-56124, Pisa, Italy.,Department of Neuroscience, Psychology, Drug Research and Child Health NEUROFARBA, University of Florence, Area San Salvi - Pad. 26, I-50135, Florence, Italy
| | - Laura Baroncelli
- Institute of Neuroscience, National Research Council (CNR), Via Moruzzi 1, I-56124, Pisa, Italy.
| | - Gabriele Sansevero
- Institute of Neuroscience, National Research Council (CNR), Via Moruzzi 1, I-56124, Pisa, Italy.,Department of Neuroscience, Psychology, Drug Research and Child Health NEUROFARBA, University of Florence, Area San Salvi - Pad. 26, I-50135, Florence, Italy
| | - Tatjana Begenisic
- Institute of Neuroscience, National Research Council (CNR), Via Moruzzi 1, I-56124, Pisa, Italy
| | - Concetta Prontera
- Fondazione G. Monasterio CNR-Regione Toscana, via Moruzzi 1, I-56124, Pisa, Italy
| | - Alessandro Sale
- Institute of Neuroscience, National Research Council (CNR), Via Moruzzi 1, I-56124, Pisa, Italy
| | - Maria Cristina Cenni
- Institute of Neuroscience, National Research Council (CNR), Via Moruzzi 1, I-56124, Pisa, Italy
| | - Nicoletta Berardi
- Institute of Neuroscience, National Research Council (CNR), Via Moruzzi 1, I-56124, Pisa, Italy.,Department of Neuroscience, Psychology, Drug Research and Child Health NEUROFARBA, University of Florence, Area San Salvi - Pad. 26, I-50135, Florence, Italy
| | - Lamberto Maffei
- Institute of Neuroscience, National Research Council (CNR), Via Moruzzi 1, I-56124, Pisa, Italy
| |
Collapse
|
25
|
Lee A, Shen M, Qiu A. Psychiatric polygenic risk associates with cortical morphology and functional organization in aging. Transl Psychiatry 2017; 7:1276. [PMID: 29225336 PMCID: PMC5802582 DOI: 10.1038/s41398-017-0036-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 08/04/2017] [Accepted: 09/07/2017] [Indexed: 01/23/2023] Open
Abstract
Common brain abnormalities in cortical morphology and functional organization are observed in psychiatric disorders and aging, reflecting shared genetic influences. This preliminary study aimed to examine the contribution of a polygenetic risk for psychiatric disorders (PRScross) to aging brain and to identify molecular mechanisms through the use of multimodal brain images, genotypes, and transcriptome data. We showed age-related cortical thinning in bilateral inferior frontal cortex (IFC) and superior temporal gyrus and alterations in the functional connectivity between bilateral IFC and between right IFC and right inferior parietal lobe as a function of PRScross. Interestingly, the genes in PRScross, that contributed most to aging neurodegeneration, were expressed in the functioanlly connected cortical regions. Especially, genes identified through the genotype-functional connectivity association analysis were commonly expressed in both cortical regions and formed strong gene networks with biological processes related to neural plasticity and synaptogenesis, regulated by glutamatergic and GABAergic transmission, neurotrophin signaling, and metabolism. This study suggested integrating genotype and transcriptome with neuroimage data sheds new light on the mechanisms of aging brain.
Collapse
Affiliation(s)
- Annie Lee
- 0000 0001 2180 6431grid.4280.eDepartment of Biomedical Engineering, National University of Singapore, Singapore, 117576 Singapore
| | - Mojun Shen
- 0000 0004 0637 0221grid.185448.4Singapore Institute for Clinical Sciences, The Agency for Science, Technology and Research, Singapore, 117609 Singapore
| | - Anqi Qiu
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore. .,Singapore Institute for Clinical Sciences, The Agency for Science, Technology and Research, Singapore, 117609, Singapore. .,Clinical Imaging Research Center, National University of Singapore, Singapore, 117456, Singapore.
| |
Collapse
|
26
|
Tian Z, Zhao Q, Biswas S, Deng W. Methods of reactivation and reprogramming of neural stem cells for neural repair. Methods 2017; 133:3-20. [PMID: 28864354 DOI: 10.1016/j.ymeth.2017.08.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 07/21/2017] [Accepted: 08/24/2017] [Indexed: 12/27/2022] Open
Abstract
Research on the biology of adult neural stem cells (NSCs) and induced NSCs (iNSCs), as well as NSC-based therapies for diseases in central nervous system (CNS) has started to generate the expectation that these cells may be used for treatments in CNS injuries or disorders. Recent technological progresses in both NSCs themselves and their derivatives have brought us closer to therapeutic applications. Adult neurogenesis presents in particular regions in mammal brain, known as neurogenic niches such as the dental gyrus (DG) in hippocampus and the subventricular zone (SVZ), within which adult NSCs usually stay for long periods out of the cell cycle, in G0. The reactivation of quiescent adult NSCs needs orchestrated interactions between the extrinsic stimulis from niches and the intrinsic factors involving transcription factors (TFs), signaling pathway, epigenetics, and metabolism to start an intracellular regulatory program, which promotes the quiescent NSCs exit G0 and reenter cell cycle. Extrinsic and intrinsic mechanisms that regulate adult NSCs are interconnected and feedback on one another. Since endogenous neurogenesis only happens in restricted regions and steadily fails with disease advances, interest has evolved to apply the iNSCs converted from somatic cells to treat CNS disorders, as is also promising and preferable. To overcome the limitation of viral-based reprogramming of iNSCs, bioactive small molecules (SM) have been explored to enhance the efficiency of iNSC reprogramming or even replace TFs, making the iNSCs more amenable to clinical application. Despite intense research efforts to translate the studies of adult and induced NSCs from the bench to bedside, vital troubles remain at several steps in these processes. In this review, we examine the present status, advancement, pitfalls, and potential of the two types of NSC technologies, focusing on each aspects of reactivation of quiescent adult NSC and reprogramming of iNSC from somatic cells, as well as on progresses in cell-based regenerative strategies for neural repair and criteria for successful therapeutic applications.
Collapse
Affiliation(s)
- Zuojun Tian
- Department of Neurology, The Institute of Guangzhou Respiratory Disease, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, PR China; Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, CA 95817, USA; Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, USA
| | - Qiuge Zhao
- Department of Neurology, The Institute of Guangzhou Respiratory Disease, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, PR China
| | - Sangita Biswas
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, CA 95817, USA; Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, USA.
| | - Wenbin Deng
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, CA 95817, USA; Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, USA.
| |
Collapse
|
27
|
IGF-1 mediated Neurogenesis Involves a Novel RIT1/Akt/Sox2 Cascade. Sci Rep 2017; 7:3283. [PMID: 28607354 PMCID: PMC5468318 DOI: 10.1038/s41598-017-03641-9] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 05/02/2017] [Indexed: 12/20/2022] Open
Abstract
Insulin-like growth factor 1 (IGF-1) is known to have diverse effects on brain structure and function, including the promotion of stem cell proliferation and neurogenesis in the adult dentate gyrus. However, the intracellular pathways downstream of the IGF-1 receptor that contribute to these diverse physiological actions remain relatively uncharacterized. Here, we demonstrate that the Ras-related GTPase, RIT1, plays a critical role in IGF-1-dependent neurogenesis. Studies in hippocampal neuronal precursor cells (HNPCs) demonstrate that IGF-1 stimulates a RIT1-dependent increase in Sox2 levels, resulting in pro-neural gene expression and increased cellular proliferation. In this novel cascade, RIT1 stimulates Akt-dependent phosphorylation of Sox2 at T118, leading to its stabilization and transcriptional activation. When compared to wild-type HNPCs, RIT1−/− HNPCs show deficient IGF-1-dependent Akt signaling and neuronal differentiation, and accordingly, Sox2-dependent hippocampal neurogenesis is significantly blunted following IGF-1 infusion in knockout (RIT1−/−) mice. Consistent with a role for RIT1 function in the modulation of activity-dependent plasticity, exercise-mediated potentiation of hippocampal neurogenesis is also diminished in RIT1−/− mice. Taken together, these data identify the previously uncharacterized IGF1-RIT1-Akt-Sox2 signaling pathway as a key component of neurogenic niche sensing, contributing to the regulation of neural stem cell homeostasis.
Collapse
|
28
|
Long Q, Luo Q, Wang K, Bates A, Shetty AK. Mash1-dependent Notch Signaling Pathway Regulates GABAergic Neuron-Like Differentiation from Bone Marrow-Derived Mesenchymal Stem Cells. Aging Dis 2017; 8:301-313. [PMID: 28580186 PMCID: PMC5440110 DOI: 10.14336/ad.2016.1018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 10/18/2016] [Indexed: 12/16/2022] Open
Abstract
GABAergic neuronal cell grafting has promise for treating a multitude of neurological disorders including epilepsy, age-related memory dysfunction, Alzheimer's disease and schizophrenia. However, identification of an unlimited source of GABAergic cells is critical for advancing such therapies. Our previous study implied that reprogramming of bone marrow-derived mesenchymal stem cells (BMSCs) through overexpression of the Achaete-scute homolog 1 (Ascl1, also called Mash1) could generate GABAergic neuron-like cells. Here, we investigated mechanisms underlying the conversion of BMSCs into GABAergic cells. We inhibited γ-secretase (an enzyme that activates Notch signaling) with N-[N-(3,5-difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester (DAPT) or manipulated the expression of Notch signaling components such as the recombination signal binding protein for immunoglobulin kappa J region (RBPJ), hairy and enhancer of split-1 (Hes1) or Mash1. We demonstrate that inhibition of γ-secretase through DAPT down-regulates RBPJ and Hes1, up-regulates Mash1 and results in an enhanced differentiation of BMSCs into GABAergic cells. On the other hand, RBPJ knockdown in BMSCs has no effect on Mash1 gene expression whereas Hes1 knockdown increases the expression of Mash1. Transduction of Mash1 in BMSCs also increases the expression of Hes1 but not RBPJ. Moreover, increased GABAergic differentiation in BMSCs occurs with concurrent Mash1 overexpression and Hes1-silencing. Thus, the Mash1-dependent Notch signaling pathway regulates GABAergic neuron-like differentiation of BMSCs. These results also suggest that genetic engineering of BMSCs is a useful avenue for obtaining GABAergic neuron-like donor cells for the treatment of neurological disorders.
Collapse
Affiliation(s)
- Qianfa Long
- 1Department of Neurosurgery, Xi'an Central Hospital, Xi'an Jiao Tong University School of Medicine, Xi'an 710003, China.,2Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine at Scott & White, Temple and College Station, Texas, 76502, USA
| | - Qiang Luo
- 1Department of Neurosurgery, Xi'an Central Hospital, Xi'an Jiao Tong University School of Medicine, Xi'an 710003, China
| | - Kai Wang
- 3Department of Neurosurgery, Qingdao 401 Hospital of PLA, Qingdao 266071, China
| | - Adrian Bates
- 2Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine at Scott & White, Temple and College Station, Texas, 76502, USA.,4Research Service, Olin E. Teague Veterans' Medical Center, CTVHCS, Temple, Texas, USA
| | - Ashok K Shetty
- 2Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine at Scott & White, Temple and College Station, Texas, 76502, USA.,4Research Service, Olin E. Teague Veterans' Medical Center, CTVHCS, Temple, Texas, USA
| |
Collapse
|
29
|
Bryan MR, Bowman AB. Manganese and the Insulin-IGF Signaling Network in Huntington's Disease and Other Neurodegenerative Disorders. ADVANCES IN NEUROBIOLOGY 2017; 18:113-142. [PMID: 28889265 PMCID: PMC6559248 DOI: 10.1007/978-3-319-60189-2_6] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disease resulting in motor impairment and death in patients. Recently, several studies have demonstrated insulin or insulin-like growth factor (IGF) treatment in models of HD, resulting in potent amelioration of HD phenotypes via modulation of the PI3K/AKT/mTOR pathways. Administration of IGF and insulin can rescue microtubule transport, metabolic function, and autophagy defects, resulting in clearance of Huntingtin (HTT) aggregates, restoration of mitochondrial function, amelioration of motor abnormalities, and enhanced survival. Manganese (Mn) is an essential metal to all biological systems but, in excess, can be toxic. Interestingly, several studies have revealed the insulin-mimetic effects of Mn-demonstrating Mn can activate several of the same metabolic kinases and increase peripheral and neuronal insulin and IGF-1 levels in rodent models. Separate studies have shown mouse and human striatal neuroprogenitor cell (NPC) models exhibit a deficit in cellular Mn uptake, indicative of a Mn deficiency. Furthermore, evidence from the literature reveals a striking overlap between cellular consequences of Mn deficiency (i.e., impaired function of Mn-dependent enzymes) and known HD endophenotypes including excitotoxicity, increased reactive oxygen species (ROS) accumulation, and decreased mitochondrial function. Here we review published evidence supporting a hypothesis that (1) the potent effect of IGF or insulin treatment on HD models, (2) the insulin-mimetic effects of Mn, and (3) the newly discovered Mn-dependent perturbations in HD may all be functionally related. Together, this review will present the intriguing possibility that intricate regulatory cross-talk exists between Mn biology and/or toxicology and the insulin/IGF signaling pathways which may be deeply connected to HD pathology and, perhaps, other neurodegenerative diseases (NDDs) and other neuropathological conditions.
Collapse
Affiliation(s)
- Miles R Bryan
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
- Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
| | - Aaron B Bowman
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Center in Molecular Toxicology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| |
Collapse
|
30
|
Chang CY, Chen YW, Wang TW, Lai WS. Akting up in the GABA hypothesis of schizophrenia: Akt1 deficiency modulates GABAergic functions and hippocampus-dependent functions. Sci Rep 2016; 6:33095. [PMID: 27615800 PMCID: PMC5018883 DOI: 10.1038/srep33095] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 08/19/2016] [Indexed: 12/29/2022] Open
Abstract
Accumulating evidence implies that both AKT1 and GABAA receptor (GABAAR) subunit genes are involved in schizophrenia pathogenesis. Activated Akt promotes GABAergic neuron differentiation and increases GABAAR expression on the plasma membrane. To elucidate the role of Akt1 in modulating GABAergic functions and schizophrenia-related cognitive deficits, a set of 6 in vitro and in vivo experiments was conducted. First, an Akt1/2 inhibitor was applied to evaluate its effect on GABAergic neuron-like cell formation from P19 cells. Inhibiting Akt resulted in a reduction in parvalbumin-positive neuron-like cells. In Akt1−/− and wild-type mice, seizures induced using pentylenetetrazol (a GABAAR antagonist) were measured, and GABAAR expression and GABAergic interneuron abundance in the brain were examined. Female Akt1−/− mice, but not male Akt1−/− mice, exhibited less pentylenetetrazol-induced convulsive activity than their corresponding wild-type controls. Reduced parvalbumin-positive interneuron abundance and GABAAR subunit expression, especially in the hippocampus, were also observed in female Akt1−/− mice compared to female wild-type mice. Neuromorphometric analyses revealed significantly reduced neurite complexity in hippocampal pyramidal neurons. Additionally, female Akt1−/− mice displayed increased hippocampal oscillation power and impaired spatial memory compared to female wild-type mice. Our findings suggest that Akt1 deficiency modulates GABAergic interneurons and GABAAR expression, contributing to hippocampus-dependent cognitive functional impairment.
Collapse
Affiliation(s)
- Chia-Yuan Chang
- Department of Psychology, National Taiwan University, Taipei, Taiwan
| | - Yi-Wen Chen
- Department of Psychology, National Taiwan University, Taipei, Taiwan
| | - Tsu-Wei Wang
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Wen-Sung Lai
- Department of Psychology, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Brain and Mind Sciences, National Taiwan University, Taipei, Taiwan.,Neurobiology and Cognitive Science Center, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
31
|
Dyer AH, Vahdatpour C, Sanfeliu A, Tropea D. The role of Insulin-Like Growth Factor 1 (IGF-1) in brain development, maturation and neuroplasticity. Neuroscience 2016; 325:89-99. [DOI: 10.1016/j.neuroscience.2016.03.056] [Citation(s) in RCA: 192] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 02/29/2016] [Accepted: 03/24/2016] [Indexed: 02/07/2023]
|
32
|
PDK1-Akt pathway regulates radial neuronal migration and microtubules in the developing mouse neocortex. Proc Natl Acad Sci U S A 2016; 113:E2955-64. [PMID: 27170189 DOI: 10.1073/pnas.1516321113] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Neurons migrate a long radial distance by a process known as locomotion in the developing mammalian neocortex. During locomotion, immature neurons undergo saltatory movement along radial glia fibers. The molecular mechanisms that regulate the speed of locomotion are largely unknown. We now show that the serine/threonine kinase Akt and its activator phosphoinositide-dependent protein kinase 1 (PDK1) regulate the speed of locomotion of mouse neocortical neurons through the cortical plate. Inactivation of the PDK1-Akt pathway impaired the coordinated movement of the nucleus and centrosome, a microtubule-dependent process, during neuronal migration. Moreover, the PDK1-Akt pathway was found to control microtubules, likely by regulating the binding of accessory proteins including the dynactin subunit p150(glued) Consistent with this notion, we found that PDK1 regulates the expression of cytoplasmic dynein intermediate chain and light intermediate chain at a posttranscriptional level in the developing neocortex. Our results thus reveal an essential role for the PDK1-Akt pathway in the regulation of a key step of neuronal migration.
Collapse
|
33
|
The glucagon-like peptide 1 (GLP) receptor as a therapeutic target in Parkinson's disease: mechanisms of action. Drug Discov Today 2016; 21:802-18. [PMID: 26851597 DOI: 10.1016/j.drudis.2016.01.013] [Citation(s) in RCA: 260] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 12/03/2015] [Accepted: 01/25/2016] [Indexed: 02/06/2023]
|
34
|
Oishi K, Nakagawa N, Tachikawa K, Sasaki S, Aramaki M, Hirano S, Yamamoto N, Yoshimura Y, Nakajima K. Identity of neocortical layer 4 neurons is specified through correct positioning into the cortex. eLife 2016; 5. [PMID: 26880563 PMCID: PMC4764574 DOI: 10.7554/elife.10907] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 01/28/2016] [Indexed: 11/13/2022] Open
Abstract
Many cell-intrinsic mechanisms have been shown to regulate neuronal subtype specification in the mammalian neocortex. However, how much cell environment is crucial for subtype determination still remained unclear. Here, we show that knockdown of Protocadherin20 (Pcdh20), which is expressed in post-migratory neurons of layer 4 (L4) lineage, caused the cells to localize in L2/3. The ectopically positioned "future L4 neurons" lost their L4 characteristics but acquired L2/3 characteristics. Knockdown of a cytoskeletal protein in the future L4 neurons, which caused random disruption of positioning, also showed that those accidentally located in L4 acquired the L4 characteristics. Moreover, restoration of positioning of the Pcdh20-knockdown neurons into L4 rescued the specification failure. We further suggest that the thalamocortical axons provide a positional cue to specify L4 identity. These results suggest that the L4 identity is not completely determined at the time of birth but ensured by the surrounding environment after appropriate positioning.
Collapse
Affiliation(s)
- Koji Oishi
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Nao Nakagawa
- Division of Visual Information Processing, National Institute for Physiological Sciences, National Institutes for Natural Sciences, Okazaki, Japan.,Department of Physiological Sciences, Graduate University for Advanced Studies, Okazaki, Japan
| | - Kashiko Tachikawa
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Shinji Sasaki
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Michihiko Aramaki
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Shinji Hirano
- Department of Cell Biology, Kansai Medical University, Osaka, Japan
| | - Nobuhiko Yamamoto
- Laboratory of Cellular and Molecular Neurobiology, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Yumiko Yoshimura
- Division of Visual Information Processing, National Institute for Physiological Sciences, National Institutes for Natural Sciences, Okazaki, Japan.,Department of Physiological Sciences, Graduate University for Advanced Studies, Okazaki, Japan
| | - Kazunori Nakajima
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
35
|
Abstract
It has long been established that the transcriptional activity of retinoic acid (RA) is mediated by members of the nuclear receptor family of ligand-activated transcription factors termed RA receptors (RARs). More recent observations have established that RA also activates an additional nuclear receptor, PPARβ/δ. Partitioning RA between RARs and PPARβ/δ is governed by different intracellular lipid-binding proteins: cellular RA binding protein 2 (CRABP2) delivers RA to nuclear RARs and a fatty acid binding protein (FABP5) delivers the hormone from the cytosol to nuclear PPARβ/δ. Consequently, RA signals through RARs in CRABP2-expressing cells, but activates PPARβ/δ in cells that express a high level of FABP5. RA elicits different and sometimes opposing responses in cells that express different FABP5/CRABP2 ratios because PPARβ/δ and RARs regulate the expression of distinct sets of genes. An overview of the observations that led to the discovery of this non-classical activity of RA are presented here, along with a discussion of evidence demonstrating the involvement of the dual transcriptional activities of RA in regulating energy homeostasis, insulin responses, and adipocyte and neuron differentiation.
Collapse
|
36
|
Forkhead box O transcription factors as possible mediators in the development of major depression. Neuropharmacology 2015; 99:527-37. [DOI: 10.1016/j.neuropharm.2015.08.020] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 07/22/2015] [Accepted: 08/12/2015] [Indexed: 01/26/2023]
|
37
|
Fukuzaki Y, Shin H, Kawai HD, Yamanoha B, Kogure S. 532 nm Low-Power Laser Irradiation Facilitates the Migration of GABAergic Neural Stem/Progenitor Cells in Mouse Neocortex. PLoS One 2015; 10:e0123833. [PMID: 25919297 PMCID: PMC4412395 DOI: 10.1371/journal.pone.0123833] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 02/21/2015] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND AND OBJECTIVE Accumulating evidence has shown that low-power laser irradiation (LLI) affects cell proliferation and survival, but little is known about LLI effects on neural stem/progenitor cells (NSPCs). Here we investigate whether transcranial 532 nm LLI affects NSPCs in adult murine neocortex and in neurospheres from embryonic mice. STUDY DESIGN/MATERIALS AND METHODS We applied 532 nm LLI (Nd:YVO4, CW, 60 mW) on neocortical surface via cranium in adult mice and on cultured cells from embryonic mouse brains in vitro to investigate the proliferation and migration of NSPCs and Akt expression using immunohistochemical assays and Western blotting techniques. RESULTS In vivo experiments demonstrated that 532 nm LLI significantly facilitated the migration of GABAergic NSPCs that were induced to proliferate in layer 1 by mild ischemia. In vitro experiments using GABAergic NSPCs derived from embryonic day 14 ganglionic eminence demonstrated that 532 nm LLI for 60 min promoted the migration of GAD67-immunopositive NSPCs with a significant increase of Akt expression. Meanwhile, the LLI induced proliferation, but not migration, of NSPCs that give rise to excitatory neurons. CONCLUSION It is concluded that 532 nm LLI promoted the migration of GABAergic NSPCs into deeper layers of the neocortex in vivo by elevating Akt expression.
Collapse
Affiliation(s)
- Yumi Fukuzaki
- Department of Bioinformatics, Graduate School of Engineering, Soka University, Hachioji, Tokyo, Japan
| | - Hyeryun Shin
- Department of Bioinformatics, Graduate School of Engineering, Soka University, Hachioji, Tokyo, Japan
| | - Hideki D. Kawai
- Department of Bioinformatics, Graduate School of Engineering, Soka University, Hachioji, Tokyo, Japan
| | - Banri Yamanoha
- Department of Environmental Engineering for Symbiosis, Faculty of Engineering, Soka University, Hachioji, Tokyo, Japan
| | - Shinichi Kogure
- Department of Bioinformatics, Graduate School of Engineering, Soka University, Hachioji, Tokyo, Japan
- * E-mail:
| |
Collapse
|
38
|
Genin EC, Caron N, Vandenbosch R, Nguyen L, Malgrange B. Concise review: forkhead pathway in the control of adult neurogenesis. Stem Cells 2015; 32:1398-407. [PMID: 24510844 DOI: 10.1002/stem.1673] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 01/09/2014] [Accepted: 01/09/2014] [Indexed: 12/23/2022]
Abstract
New cells are continuously generated from immature proliferating cells in the adult brain in two neurogenic niches known as the subgranular zone (SGZ) of the dentate gyrus (DG) of the hippocampus and the sub-ventricular zone (SVZ) of the lateral ventricles. However, the molecular mechanisms regulating their proliferation, differentiation, migration and functional integration of newborn neurons in pre-existing neural network remain largely unknown. Forkhead box (Fox) proteins belong to a large family of transcription factors implicated in a wide variety of biological processes. Recently, there has been accumulating evidence that several members of this family of proteins play important roles in adult neurogenesis. Here, we describe recent advances in our understanding of regulation provided by Fox factors in adult neurogenesis, and evaluate the potential role of Fox proteins as targets for therapeutic intervention in neurodegenerative diseases.
Collapse
Affiliation(s)
- Emmanuelle C Genin
- GIGA-Neurosciences, Developmental Neurobiology Unit, University of Liège, Liège, Belgium
| | | | | | | | | |
Collapse
|
39
|
Vasconcelos FF, Castro DS. Transcriptional control of vertebrate neurogenesis by the proneural factor Ascl1. Front Cell Neurosci 2014; 8:412. [PMID: 25520623 PMCID: PMC4251449 DOI: 10.3389/fncel.2014.00412] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Accepted: 11/12/2014] [Indexed: 11/19/2022] Open
Abstract
Proneural transcription factors (TFs) such as Ascl1 function as master regulators of neurogenesis in vertebrates, being both necessary and sufficient for the activation of a full program of neuronal differentiation. Novel insights into the dynamics of Ascl1 expression at the cellular level, combined with the progressive characterization of its transcriptional program, have expanded the classical view of Ascl1 as a differentiation factor in neurogenesis. These advances resulted in a new model, whereby Ascl1 promotes sequentially the proliferation and differentiation of neural/stem progenitor cells. The multiple activities of Ascl1 are associated with the activation of distinct direct targets at progressive stages along the neuronal lineage. How this temporal pattern is established is poorly understood. Two modes of Ascl1 expression recently described (oscillatory vs. sustained) are likely to be of importance, together with additional mechanistic determinants such as the chromatin landscape and other transcriptional pathways. Here we revise these latest findings, and discuss their implications to the gene regulatory functions of Ascl1 during neurogenesis.
Collapse
Affiliation(s)
| | - Diogo S Castro
- Molecular Neurobiology, Instituto Gulbenkian de Ciência Oeiras, Portugal
| |
Collapse
|
40
|
Urbán N, Guillemot F. Neurogenesis in the embryonic and adult brain: same regulators, different roles. Front Cell Neurosci 2014; 8:396. [PMID: 25505873 PMCID: PMC4245909 DOI: 10.3389/fncel.2014.00396] [Citation(s) in RCA: 352] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 11/05/2014] [Indexed: 12/12/2022] Open
Abstract
Neurogenesis persists in adult mammals in specific brain areas, known as neurogenic niches. Adult neurogenesis is highly dynamic and is modulated by multiple physiological stimuli and pathological states. There is a strong interest in understanding how this process is regulated, particularly since active neuronal production has been demonstrated in both the hippocampus and the subventricular zone (SVZ) of adult humans. The molecular mechanisms that control neurogenesis have been extensively studied during embryonic development. Therefore, we have a broad knowledge of the intrinsic factors and extracellular signaling pathways driving proliferation and differentiation of embryonic neural precursors. Many of these factors also play important roles during adult neurogenesis, but essential differences exist in the biological responses of neural precursors in the embryonic and adult contexts. Because adult neural stem cells (NSCs) are normally found in a quiescent state, regulatory pathways can affect adult neurogenesis in ways that have no clear counterpart during embryogenesis. BMP signaling, for instance, regulates NSC behavior both during embryonic and adult neurogenesis. However, this pathway maintains stem cell proliferation in the embryo, while it promotes quiescence to prevent stem cell exhaustion in the adult brain. In this review, we will compare and contrast the functions of transcription factors (TFs) and other regulatory molecules in the embryonic brain and in adult neurogenic regions of the adult brain in the mouse, with a special focus on the hippocampal niche and on the regulation of the balance between quiescence and activation of adult NSCs in this region.
Collapse
Affiliation(s)
- Noelia Urbán
- Department of Molecular Neurobiology, MRC National Institute for Medical Research London, UK
| | - François Guillemot
- Department of Molecular Neurobiology, MRC National Institute for Medical Research London, UK
| |
Collapse
|
41
|
A transcriptional mechanism integrating inputs from extracellular signals to activate hippocampal stem cells. Neuron 2014; 83:1085-97. [PMID: 25189209 PMCID: PMC4157576 DOI: 10.1016/j.neuron.2014.08.004] [Citation(s) in RCA: 158] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/01/2014] [Indexed: 12/27/2022]
Abstract
The activity of adult stem cells is regulated by signals emanating from the surrounding tissue. Many niche signals have been identified, but it is unclear how they influence the choice of stem cells to remain quiescent or divide. Here we show that when stem cells of the adult hippocampus receive activating signals, they first induce the expression of the transcription factor Ascl1 and only subsequently exit quiescence. Moreover, lowering Ascl1 expression reduces the proliferation rate of hippocampal stem cells, and inactivating Ascl1 blocks quiescence exit completely, rendering them unresponsive to activating stimuli. Ascl1 promotes the proliferation of hippocampal stem cells by directly regulating the expression of cell-cycle regulatory genes. Ascl1 is similarly required for stem cell activation in the adult subventricular zone. Our results support a model whereby Ascl1 integrates inputs from both stimulatory and inhibitory signals and converts them into a transcriptional program activating adult neural stem cells.
Collapse
|
42
|
So EY, Ouchi T. Translational initiation regulated by ATM in dendritic cells development. Cell Death Dis 2014; 5:e1418. [PMID: 25210801 PMCID: PMC4540194 DOI: 10.1038/cddis.2014.362] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 06/24/2014] [Accepted: 07/01/2014] [Indexed: 01/15/2023]
Abstract
Ataxia telangiectasia mutated (ATM) protein has been implicated in multiple pathways such as DNA repair, cell cycle checkpoint, cell growth, development, and stem cell renewal. In this study, we demonstrate evidence that ATM is involved in granulocyte macrophage colony-stimulating factor (GM-CSF)-induced dendritic cell (DC) development from bone marrow (BM) cells. Inactivation of ATM protein results in decreased BM proliferation, leading to reduced DC development and their activity for T cell activation. Expression of Jak2, STAT5, and mTOR is suppressed in both wild-type and ATM-null BM prior to GM-CSF stimulation. Activation of those proteins is delayed and prolonged hypophosphorylation of 4EBP1 is observed in ATM-null BM when treated with GM-CSF, although Erk and p38 are similarly expressed and activated in both wild-type and ATM-null BM cell types. Akt is also suppressed in wild-type BM, and transduction of constitutively active Akt or STAT5 in ATM-null BM restores DC development. Together, these results illustrate that ATM deficiency causes impaired initiation of protein translation in BM, leading to immature development of DC.
Collapse
Affiliation(s)
- E Y So
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - T Ouchi
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, NY, USA
| |
Collapse
|
43
|
Kim DY, Rhee I, Paik J. Metabolic circuits in neural stem cells. Cell Mol Life Sci 2014; 71:4221-41. [PMID: 25037158 DOI: 10.1007/s00018-014-1686-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 06/25/2014] [Accepted: 07/14/2014] [Indexed: 12/15/2022]
Abstract
Metabolic activity indicative of cellular demand is emerging as a key player in cell fate decision. Numerous studies have demonstrated that diverse metabolic pathways have a critical role in the control of the proliferation, differentiation and quiescence of stem cells. The identification of neural stem/progenitor cells (NSPCs) and the characterization of their development and fate decision process have provided insight into the regenerative potential of the adult brain. As a result, the potential of NSPCs in cell replacement therapies for neurological diseases is rapidly growing. The aim of this review is to discuss the recent findings on the crosstalk among key regulators of NSPC development and the metabolic regulation crucial for the function and cell fate decisions of NSPCs. Fundamental understanding of the metabolic circuits in NSPCs may help to provide novel approaches for reactivating neurogenesis to treat degenerative brain conditions and cognitive decline.
Collapse
Affiliation(s)
- Do-Yeon Kim
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York City, NY, 10065, USA
| | | | | |
Collapse
|
44
|
Agis-Balboa RC, Fischer A. Generating new neurons to circumvent your fears: the role of IGF signaling. Cell Mol Life Sci 2014; 71:21-42. [PMID: 23543251 PMCID: PMC11113432 DOI: 10.1007/s00018-013-1316-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Revised: 02/12/2013] [Accepted: 03/04/2013] [Indexed: 12/13/2022]
Abstract
Extinction of fear memory is a particular form of cognitive function that is of special interest because of its involvement in the treatment of anxiety and mood disorders. Based on recent literature and our previous findings (EMBO J 30(19):4071-4083, 2011), we propose a new hypothesis that implies a tight relationship among IGF signaling, adult hippocampal neurogenesis and fear extinction. Our proposed model suggests that fear extinction-induced IGF2/IGFBP7 signaling promotes the survival of neurons at 2-4 weeks old that would participate in the discrimination between the original fear memory trace and the new safety memory generated during fear extinction. This is also called "pattern separation", or the ability to distinguish similar but different cues (e.g., context). To understand the molecular mechanisms underlying fear extinction is therefore of great clinical importance.
Collapse
Affiliation(s)
- R C Agis-Balboa
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Grisebach Str. 5, 37077, Göttingen, Germany,
| | | |
Collapse
|
45
|
Annenkov A. Receptor tyrosine kinase (RTK) signalling in the control of neural stem and progenitor cell (NSPC) development. Mol Neurobiol 2013; 49:440-71. [PMID: 23982746 DOI: 10.1007/s12035-013-8532-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 08/09/2013] [Indexed: 01/04/2023]
Abstract
Important developmental responses are elicited in neural stem and progenitor cells (NSPC) by activation of the receptor tyrosine kinases (RTK), including the fibroblast growth factor receptors, epidermal growth factor receptor, platelet-derived growth factor receptors and insulin-like growth factor receptor (IGF1R). Signalling through these RTK is necessary and sufficient for driving a number of developmental processes in the central nervous system. Within each of the four RTK families discussed here, receptors are activated by sets of ligands that do not cross-activate receptors of the other three families, and therefore, their activation can be independently regulated by ligand availability. These RTK pathways converge on a conserved core of signalling molecules, but differences between the receptors in utilisation of signalling molecules and molecular adaptors for intracellular signal propagation become increasingly apparent. Intracellular inhibitors of RTK signalling are widely involved in the regulation of developmental signalling in NSPC and often determine developmental outcomes of RTK activation. In addition, cellular responses of NSPC to the activation of a given RTK may be significantly modulated by signal strength. Cellular propensity to respond also plays a role in developmental outcomes of RTK signalling. In combination, these mechanisms regulate the balance between NSPC maintenance and differentiation during development and in adulthood. Attribution of particular developmental responses of NSPC to specific pathways of RTK signalling becomes increasingly elucidated. Co-activation of several RTK in developing NSPC is common, and analysis of co-operation between their signalling pathways may advance knowledge of RTK role in NSPC development.
Collapse
Affiliation(s)
- Alexander Annenkov
- Bone and Joint Research Unit, William Harvey Research Institute, Bart's and The London School of Medicine, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK,
| |
Collapse
|
46
|
Webb AE, Pollina EA, Vierbuchen T, Urbán N, Ucar D, Leeman DS, Martynoga B, Sewak M, Rando TA, Guillemot F, Wernig M, Brunet A. FOXO3 shares common targets with ASCL1 genome-wide and inhibits ASCL1-dependent neurogenesis. Cell Rep 2013; 4:477-91. [PMID: 23891001 DOI: 10.1016/j.celrep.2013.06.035] [Citation(s) in RCA: 123] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 05/11/2013] [Accepted: 06/25/2013] [Indexed: 12/16/2022] Open
Abstract
FOXO transcription factors are central regulators of longevity from worms to humans. FOXO3, the FOXO isoform associated with exceptional human longevity, preserves adult neural stem cell pools. Here, we identify FOXO3 direct targets genome-wide in primary cultures of adult neural progenitor cells (NPCs). Interestingly, FOXO3-bound sites are enriched for motifs for bHLH transcription factors, and FOXO3 shares common targets with the proneuronal bHLH transcription factor ASCL1/MASH1 in NPCs. Analysis of the chromatin landscape reveals that FOXO3 and ASCL1 are particularly enriched at the enhancers of genes involved in neurogenic pathways. Intriguingly, FOXO3 inhibits ASCL1-dependent neurogenesis in NPCs and direct neuronal conversion in fibroblasts. FOXO3 also restrains neurogenesis in vivo. Our study identifies a genome-wide interaction between the prolongevity transcription factor FOXO3 and the cell-fate determinant ASCL1 and raises the possibility that FOXO3's ability to restrain ASCL1-dependent neurogenesis may help preserve the neural stem cell pool.
Collapse
Affiliation(s)
- Ashley E Webb
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Chen YS, Hsiue GH. Directing neural differentiation of mesenchymal stem cells by carboxylated multiwalled carbon nanotubes. Biomaterials 2013; 34:4936-44. [DOI: 10.1016/j.biomaterials.2013.03.063] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Accepted: 03/25/2013] [Indexed: 12/19/2022]
|
48
|
Cheng YC, Hsieh FY, Chiang MC, Scotting PJ, Shih HY, Lin SJ, Wu HL, Lee HT. Akt1 mediates neuronal differentiation in zebrafish via a reciprocal interaction with notch signaling. PLoS One 2013; 8:e54262. [PMID: 23342113 PMCID: PMC3544797 DOI: 10.1371/journal.pone.0054262] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Accepted: 12/10/2012] [Indexed: 12/12/2022] Open
Abstract
Akt1 is well known for its role in regulating cell proliferation, differentiation, and apoptosis and is implicated in tumors and several neurological disorders. However, the role of Akt1 in neural development has not been well defined. We have isolated zebrafish akt1 and shown that this gene is primarily transcribed in the developing nervous system, and its spatiotemporal expression pattern suggests a role in neural differentiation. Injection of akt1 morpholinos resulted in loss of neuronal precursors with a concomitant increase in post-mitotic neurons, indicating that knockdown of Akt1 is sufficient to cause premature differentiation of neurons. A similar phenotype was observed in embryos deficient for Notch signaling. Both the ligand (deltaA) and the downstream target of Notch (her8a) were downregulated in akt1 morphants, indicating that Akt1 is required for Delta-Notch signaling. Furthermore, akt1 expression was downregulated in Delta-Notch signaling-deficient embryos and could be induced by constitutive activation of Notch signaling. In addition, knockdown of Akt1 was able to nullify the inhibition of neuronal differentiation caused by constitutive activation of Notch signaling. Taken together, these results provide in vivo evidence that Akt1 interacts with Notch signaling reciprocally and provide an explanation of why Akt1 is essential for the inhibition of neuronal differentiation.
Collapse
Affiliation(s)
- Yi-Chuan Cheng
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Santos T, Ferreira R, Maia J, Agasse F, Xapelli S, Cortes L, Bragança J, Malva JO, Ferreira L, Bernardino L. Polymeric nanoparticles to control the differentiation of neural stem cells in the subventricular zone of the brain. ACS NANO 2012; 6:10463-10474. [PMID: 23176155 DOI: 10.1021/nn304541h] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Herein, we report the use of retinoic acid-loaded polymeric nanoparticles as a potent tool to induce the neuronal differentiation of subventricular zone neural stem cells. The intracellular delivery of retinoic acid by the nanoparticles activated nuclear retinoic acid receptors, decreased stemness, and increased proneurogenic gene expression. Importantly, this work reports for the first time a nanoparticle formulation able to modulate in vivo the subventricular zone neurogenic niche. The work further compares the dynamics of initial stages of differentiation between SVZ cells treated with retinoic acid-loaded polymeric nanoparticles and solubilized retinoic acid. The nanoparticle formulation developed here may ultimately offer new perspectives to treat neurodegenerative diseases.
Collapse
Affiliation(s)
- Tiago Santos
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Park N, Yoo JC, Ryu J, Hong SG, Hwang EM, Park JY. Copine1 enhances neuronal differentiation of the hippocampal progenitor HiB5 cells. Mol Cells 2012; 34:549-54. [PMID: 23263657 PMCID: PMC3887833 DOI: 10.1007/s10059-012-0235-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Revised: 10/24/2012] [Accepted: 10/24/2012] [Indexed: 11/24/2022] Open
Abstract
Copine1 is a ubiquitously expressed protein found in various tissues including the brain, but little is known about the physiological function of this protein. Here, we showed that copine1 is involved in neuronal differentiation. Over-expression of copine1 clearly increased neurite outgrowth and expression of Tuj1, a neuronal marker protein, in HiB5 cells. In addition, endogenous copine1 was transiently increased at the early time during neuronal differentiation of HiB5 cells. When the expression of endogenous copine1 was knocked-down by its specific shRNA, PDGF-mediated neurite outgrowth was clearly decreased in HiB5 cells. Furthermore, over-expression of copine1 increased phosphorylation of Akt and copine1-specific shRNA decreased phosphorylation of Akt during neuronal differentiation of HiB5 cells. Interestingly, the phosphorylation level of PI3K, generally known as an upstream protein of Akt, was not changed by copine1 expression. These results suggest that copine1 enhances neuronal differentiation of HiB5 cells not through the PI3K-Akt pathway, but by using another Akt activated signal pathway.
Collapse
Affiliation(s)
- Nammi Park
- Department of Physiology, Institute of Health Science, and Medical Research Center for Neural Dysfunction, School of Medicine, Gyeongsang National University, Jinju 660-751,
Korea
| | - Jae Cheal Yoo
- Department of Physiology, Institute of Health Science, and Medical Research Center for Neural Dysfunction, School of Medicine, Gyeongsang National University, Jinju 660-751,
Korea
| | - Jiwon Ryu
- Department of Physiology, Institute of Health Science, and Medical Research Center for Neural Dysfunction, School of Medicine, Gyeongsang National University, Jinju 660-751,
Korea
| | - Seong-Geun Hong
- Department of Physiology, Institute of Health Science, and Medical Research Center for Neural Dysfunction, School of Medicine, Gyeongsang National University, Jinju 660-751,
Korea
| | - Eun Mi Hwang
- Center for Neural Science and WCI Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul 136-791,
Korea
| | - Jae-Yong Park
- Department of Physiology, Institute of Health Science, and Medical Research Center for Neural Dysfunction, School of Medicine, Gyeongsang National University, Jinju 660-751,
Korea
- Center for Neural Science and WCI Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul 136-791,
Korea
| |
Collapse
|