51
|
Guo DH, Yamamoto M, Hernandez CM, Khodadadi H, Baban B, Stranahan AM. Beige adipocytes mediate the neuroprotective and anti-inflammatory effects of subcutaneous fat in obese mice. Nat Commun 2021; 12:4623. [PMID: 34330904 PMCID: PMC8324783 DOI: 10.1038/s41467-021-24540-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 06/24/2021] [Indexed: 12/25/2022] Open
Abstract
Visceral obesity increases risk of cognitive decline in humans, but subcutaneous adiposity does not. Here, we report that beige adipocytes are indispensable for the neuroprotective and anti-inflammatory effects of subcutaneous fat. Mice lacking functional beige fat exhibit accelerated cognitive dysfunction and microglial activation with dietary obesity. Subcutaneous fat transplantation also protects against chronic obesity in wildtype mice via beige fat-dependent mechanisms. Beige adipocytes restore hippocampal synaptic plasticity following transplantation, and these effects require the anti-inflammatory cytokine interleukin-4 (IL4). After observing beige fat-mediated induction of IL4 in meningeal T-cells, we investigated the contributions of peripheral lymphocytes in donor fat. There was no sign of donor-derived lymphocyte trafficking between fat and brain, but recipient-derived lymphocytes were required for the effects of transplantation on cognition and microglial morphology. These findings indicate that beige adipocytes oppose obesity-induced cognitive impairment, with a potential role for IL4 in the relationship between beige fat and brain function.
Collapse
Affiliation(s)
- De-Huang Guo
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Masaki Yamamoto
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Caterina M Hernandez
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Hesam Khodadadi
- Department of Oral Biology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Babak Baban
- Department of Oral Biology, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Plastic Surgery Section, Department of Surgery, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Alexis M Stranahan
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA.
| |
Collapse
|
52
|
Shan S, Xu F, Brenig B. Genome-Wide Association Studies Reveal Neurological Genes for Dog Herding, Predation, Temperament, and Trainability Traits. Front Vet Sci 2021; 8:693290. [PMID: 34368281 PMCID: PMC8335642 DOI: 10.3389/fvets.2021.693290] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 06/15/2021] [Indexed: 11/13/2022] Open
Abstract
Genome-wide association study (GWAS) using dog breed standard values as phenotypic measurements is an efficient way to identify genes associated with morphological and behavioral traits. As a result of strong human purposeful selections, several specialized behavioral traits such as herding and hunting have been formed in different modern dog breeds. However, genetic analyses on this topic are rather limited due to the accurate phenotyping difficulty for these complex behavioral traits. Here, 268 dog whole-genome sequences from 130 modern breeds were used to investigate candidate genes underlying dog herding, predation, temperament, and trainability by GWAS. Behavioral phenotypes were obtained from the American Kennel Club based on dog breed standard descriptions or groups (conventional categorization of dog historical roles). The GWAS results of herding behavior (without body size as a covariate) revealed 44 significantly associated sites within five chromosomes. Significantly associated sites on CFA7, 9, 10, and 20 were located either in or near neuropathological or neuronal genes including THOC1, ASIC2, MSRB3, LLPH, RFX8, and CHL1. MSRB3 and CHL1 genes were reported to be associated with dog fear. Since herding is a restricted hunting behavior by removing killing instinct, 36 hounds and 55 herding dogs were used to analyze predation behavior. Three neuronal-related genes (JAK2, MEIS1, and LRRTM4) were revealed as candidates for predation behavior. The significantly associated variant of temperament GWAS was located within ACSS3 gene. The highest associated variant in trainability GWAS is located on CFA22, with no variants detected above the Bonferroni threshold. Since dog behaviors are correlated with body size, we next incorporate body mass as covariates into GWAS; and significant signals around THOC1, MSRB3, LLPH, RFX8, CHL1, LRRTM4, and ACSS3 genes were still detected for dog herding, predation, and temperament behaviors. In humans, these candidate genes are either involved in nervous system development or associated with mental disorders. In conclusion, our results imply that these neuronal or psychiatric genes might be involved in biological processes underlying dog herding, predation, and temperament behavioral traits.
Collapse
Affiliation(s)
- Shuwen Shan
- Department of Animal Sciences, Faculty of Agricultural Sciences, Institute of Veterinary Medicine, University of Goettingen, Göttingen, Germany
| | - Fangzheng Xu
- Department of Animal Sciences, Faculty of Agricultural Sciences, Institute of Veterinary Medicine, University of Goettingen, Göttingen, Germany
| | - Bertram Brenig
- Department of Animal Sciences, Faculty of Agricultural Sciences, Institute of Veterinary Medicine, University of Goettingen, Göttingen, Germany
| |
Collapse
|
53
|
Pan Y, He X, Li C, Li Y, Li W, Zhang H, Wang Y, Zhou G, Yang J, Li J, Qu J, Wang H, Gao Z, Shen Y, Li T, Hu H, Ma H. Neuronal activity recruits the CRTC1/CREB axis to drive transcription-dependent autophagy for maintaining late-phase LTD. Cell Rep 2021; 36:109398. [PMID: 34289350 DOI: 10.1016/j.celrep.2021.109398] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 05/05/2021] [Accepted: 06/22/2021] [Indexed: 12/31/2022] Open
Abstract
Cellular resources must be reorganized for long-term synaptic plasticity during brain information processing, in which coordinated gene transcription and protein turnover are required. However, the mechanism underlying this process remains elusive. Here, we report that activating N-methyl-d-aspartate receptors (NMDARs) induce transcription-dependent autophagy for synaptic turnover and late-phase long-term synaptic depression (L-LTD), which invokes cytoplasm-to-nucleus signaling mechanisms known to be required for late-phase long-term synaptic potentiation (L-LTP). Mechanistically, LTD-inducing stimuli specifically dephosphorylate CRTC1 (CREB-regulated transcription coactivator 1) at Ser-151 and are advantaged in recruiting CRTC1 from cytoplasm to the nucleus, where it competes with FXR (fed-state sensing nuclear receptor) for binding to CREB (cAMP response element-binding protein) and drives autophagy gene expression. Disrupting synergistic actions of CREB and CRTC1 (two essential L-LTP transcription factors) impairs transcription-dependent autophagy induction and prevents NMDAR-dependent L-LTD, which can be rescued by constitutively inducing mechanistic target of rapamycin (mTOR)-dependent autophagy. Together, these findings uncover mechanistic commonalities between L-LTP and L-LTD, suggesting that synaptic activity can tune excitation-transcription coupling for distinct long-lasting synaptic remodeling.
Collapse
Affiliation(s)
- Yue Pan
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Affiliated Mental Health Center, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xingzhi He
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Affiliated Mental Health Center, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| | - Cuicui Li
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Affiliated Mental Health Center, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yanjun Li
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Affiliated Mental Health Center, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| | - Wenwen Li
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Affiliated Mental Health Center, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| | - Hanbin Zhang
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Affiliated Mental Health Center, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yang Wang
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Affiliated Mental Health Center, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| | - Guangjun Zhou
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Affiliated Mental Health Center, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jing Yang
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Affiliated Mental Health Center, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jiarui Li
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Affiliated Mental Health Center, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jing Qu
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Affiliated Mental Health Center, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| | - Hao Wang
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Affiliated Mental Health Center, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| | - Zhihua Gao
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Affiliated Mental Health Center, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| | - Ying Shen
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Affiliated Mental Health Center, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| | - Tao Li
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Affiliated Mental Health Center, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| | - Hailan Hu
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Affiliated Mental Health Center, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China; Research Units for Emotion and Emotion Disorders, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Huan Ma
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Affiliated Mental Health Center, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China; Research Units for Emotion and Emotion Disorders, Chinese Academy of Medical Sciences, Beijing 100730, China.
| |
Collapse
|
54
|
Li P, Wu Q, Li X, Hu B, Wen W, Xu S. Shenqi Yizhi Granule attenuates Aβ 1-42 induced cognitive dysfunction via inhibiting JAK2/STAT3 activated astrocyte reactivity. Exp Gerontol 2021; 151:111400. [PMID: 33974937 DOI: 10.1016/j.exger.2021.111400] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 03/29/2021] [Accepted: 05/05/2021] [Indexed: 01/23/2023]
Abstract
Shenqi Yizhi Granule (SYG), a modern preparation herbs based on the theory of traditional Chinese medicine, has been proved to be effective against Alzheimer's disease in clinical trials, APP/PS1 mice and 5XFAD transgenic mice. But the underlying mechanism remains ambiguous. Increasing evidence supports the crucial role of astrocyte reactivity in the pathogenesis of Alzheimer's disease (AD). In the present study, we attempt to explore the underlying mechanisms of SYG from astrocyte reactivity in Aβ1-42-induced rat model of Alzheimer's disease. After SYG treatment, the impairment of learning and memory induced by Aβ1-42 was significantly improved and the hippocampal neuron damages were alleviated. Additionally, the activity of glutamine synthetase and the concentration of glutamate, which might be involved in the cognitive dysfunctions, were outstandingly reduced. Meanwhile, the astrocyte reactivity was also remarkably inhibited. The expressions of JAK2 and STAT3, key proteins in the JAK2/STAT3 signaling pathway that is tightly associated with reactive astrocytes, were clearly attenuated, too. Collectively, our data demonstrate that SYG might exert protective effects on cognitive impairment induced by amyloid-β oligomers via inhibition of astrocyte reactivity regulated by the JAK2/STAT3 signaling pathway. It may be a potential therapeutic for cognitive dysfunctions in many neurological and psychiatric disorders such as Alzheimer's disease.
Collapse
Affiliation(s)
- Ping Li
- Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Qian Wu
- Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Xiaoqiong Li
- Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Bangyan Hu
- Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Wen Wen
- Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Shijun Xu
- Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China.
| |
Collapse
|
55
|
Lyra E Silva NM, Gonçalves RA, Pascoal TA, Lima-Filho RAS, Resende EDPF, Vieira ELM, Teixeira AL, de Souza LC, Peny JA, Fortuna JTS, Furigo IC, Hashiguchi D, Miya-Coreixas VS, Clarke JR, Abisambra JF, Longo BM, Donato J, Fraser PE, Rosa-Neto P, Caramelli P, Ferreira ST, De Felice FG. Pro-inflammatory interleukin-6 signaling links cognitive impairments and peripheral metabolic alterations in Alzheimer's disease. Transl Psychiatry 2021; 11:251. [PMID: 33911072 PMCID: PMC8080782 DOI: 10.1038/s41398-021-01349-z] [Citation(s) in RCA: 137] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/25/2021] [Accepted: 03/19/2021] [Indexed: 12/22/2022] Open
Abstract
Alzheimer's disease (AD) is associated with memory impairment and altered peripheral metabolism. Mounting evidence indicates that abnormal signaling in a brain-periphery metabolic axis plays a role in AD pathophysiology. The activation of pro-inflammatory pathways in the brain, including the interleukin-6 (IL-6) pathway, comprises a potential point of convergence between memory dysfunction and metabolic alterations in AD that remains to be better explored. Using T2-weighted magnetic resonance imaging (MRI), we observed signs of probable inflammation in the hypothalamus and in the hippocampus of AD patients when compared to cognitively healthy control subjects. Pathological examination of post-mortem AD hypothalamus revealed the presence of hyperphosphorylated tau and tangle-like structures, as well as parenchymal and vascular amyloid deposits surrounded by astrocytes. T2 hyperintensities on MRI positively correlated with plasma IL-6, and both correlated inversely with cognitive performance and hypothalamic/hippocampal volumes in AD patients. Increased IL-6 and suppressor of cytokine signaling 3 (SOCS3) were observed in post-mortem AD brains. Moreover, activation of the IL-6 pathway was observed in the hypothalamus and hippocampus of AD mice. Neutralization of IL-6 and inhibition of the signal transducer and activator of transcription 3 (STAT3) signaling in the brains of AD mouse models alleviated memory impairment and peripheral glucose intolerance, and normalized plasma IL-6 levels. Collectively, these results point to IL-6 as a link between cognitive impairment and peripheral metabolic alterations in AD. Targeting pro-inflammatory IL-6 signaling may be a strategy to alleviate memory impairment and metabolic alterations in the disease.
Collapse
Affiliation(s)
- Natalia M Lyra E Silva
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
| | - Rafaella A Gonçalves
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Tharick A Pascoal
- Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Ricardo A S Lima-Filho
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Elisa de Paula França Resende
- Behavioral and Cognitive Neurology Research Group, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
- Hospital das Clínicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Erica L M Vieira
- Centre of Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Antonio L Teixeira
- Neuropsychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Santa Casa BH Ensino e Pesquisa, Belo Horizonte, MG, Brazil
| | - Leonardo C de Souza
- Behavioral and Cognitive Neurology Research Group, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
- Hospital das Clínicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Julyanna A Peny
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Juliana T S Fortuna
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Isadora C Furigo
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Debora Hashiguchi
- Department of Physiology, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Vivian S Miya-Coreixas
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Julia R Clarke
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Jose F Abisambra
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease University of Florida, Gainesville, FL, USA
| | - Beatriz M Longo
- Department of Physiology, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Jose Donato
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Paul E Fraser
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Paulo Caramelli
- Behavioral and Cognitive Neurology Research Group, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Sergio T Ferreira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - Fernanda G De Felice
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada.
- Department of Psychiatry, Queen's University, Kingston, ON, Canada.
- Department of Biomedical and Molecuar Sciences, Queen's University, Kingston, ON, Canada.
| |
Collapse
|
56
|
Irving A, Harvey J. Regulation of hippocampal synaptic function by the metabolic hormone leptin: Implications for health and disease. Prog Lipid Res 2021; 82:101098. [PMID: 33895229 DOI: 10.1016/j.plipres.2021.101098] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/16/2021] [Accepted: 04/20/2021] [Indexed: 10/21/2022]
Abstract
Significant advances have been made in our understanding of the hormone, leptin and its CNS actions in recent years. It is now evident that leptin has a multitude of brain functions, that extend beyond its established role in the hypothalamic control of energy balance. Additional brain regions including the hippocampus are important targets for leptin, with a high density of leptin receptors (LepRs) expressed in specific hippocampal regions and localised to CA1 synapses. Extensive evidence indicates that leptin has pro-cognitive actions, as it rapidly modifies synaptic efficacy at excitatory Schaffer collateral (SC)-CA1 and temporoammonic (TA)-CA1 synapses and enhances performance in hippocampal-dependent memory tasks. There is a functional decline in hippocampal responsiveness to leptin with age, with significant reductions in the modulatory effects of leptin at SC-CA1 and TA-CA1 synapses in aged, compared to adult hippocampus. As leptin has pro-cognitive effects, this decline in leptin sensitivity is likely to have negative consequences for cognitive function during the aging process. Here we review how evaluation of the hippocampal actions of leptin has improved our knowledge of the regulatory brain functions of leptin in health and provided significant insight into the impact of leptin in age-related neurodegenerative disorders linked to cognitive decline.
Collapse
Affiliation(s)
- Andrew Irving
- School of Biomolecular and Biomedical Science, The Conway Institute, University College Dublin, Dublin, Ireland
| | - Jenni Harvey
- Division of Systems Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, United Kingdom.
| |
Collapse
|
57
|
An activity-dependent determinant of synapse elimination in the mammalian brain. Neuron 2021; 109:1333-1349.e6. [PMID: 33770504 DOI: 10.1016/j.neuron.2021.03.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/26/2021] [Accepted: 03/04/2021] [Indexed: 01/06/2023]
Abstract
To establish functional neural circuits in the brain, synaptic connections are refined by neural activity during development, where active connections are maintained and inactive ones are eliminated. However, the molecular signals that regulate synapse refinement remain to be elucidated. When we inactivate a subset of neurons in the mouse cingulate cortex, their callosal connections are eliminated through activity-dependent competition. Using this system, we identify JAK2 tyrosine kinase as a key regulator of inactive synapse elimination. We show that JAK2 is necessary and sufficient for elimination of inactive connections; JAK2 is activated at inactive synapses in response to signals from other active synapses; STAT1, a substrate of JAK2, mediates inactive synapse elimination; JAK2 signaling is critical for physiological refinement of synapses during normal development; and JAK2 regulates synapse refinement in multiple brain regions. We propose that JAK2 is an activity-dependent switch that serves as a determinant of inactive synapse elimination.
Collapse
|
58
|
Wan HL, Hong XY, Zhao ZH, Li T, Zhang BG, Liu Q, Zhao S, Wang JZ, Shen XF, Liu GP, Liu GP. STAT3 ameliorates cognitive deficits via regulation of NMDAR expression in an Alzheimer's disease animal model. Am J Cancer Res 2021; 11:5511-5524. [PMID: 33859760 PMCID: PMC8039956 DOI: 10.7150/thno.56541] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 02/17/2021] [Indexed: 12/29/2022] Open
Abstract
Background: Abnormal tau accumulation in the brain has a positively correlation with neurodegeneration and memory deterioration, but the mechanism underlying tau-associated synaptic and cognitive impairments remains unclear. Our previous work has found that human full length tau (hTau) accumulation activated signal transducer and activator of transcription-1 (STAT1) to suppress N-methyl-D-aspartate receptors (NMDARs) expression, followed by memory deficits. STAT3 also belongs to STAT protein family and is reported to involve in regulation of synaptic plasticity and cognition. Here, we investigated the role of STAT3 in the cognitive deficits induced by hTau accumulation. Methods: In vitro studies HEK293 cells were used. EMSA, Luciferase reporter assay, and Immunoprecipitation were applied to detect STAT3 activity. In vivo studies, AAV virus were injected into the hippocampal CA3 region of C57 mice. Western blotting, quantitative real-time polymerase chain reaction, and immunofluorescence were applied to examine the level of synaptic proteins. Electrophysiological analysis, behavioral testing and Golgi impregnation were used to determine synaptic plasticity and memory ability recovery after overexpressing STAT3 or non-acetylated STAT1. Results: Our results showed that hTau accumulation acetylated STAT1 to retain STAT3 in the cytoplasm by increasing the binding of STAT1 with STAT3, and thus inactivated STAT3. Overexpressing STAT3 or non-acetylated STAT1 ameliorated hTau-induced synaptic loss and memory deficits by increasing the expression of NMDARs. Conclusions: Taken together, our study indicates that hTau accumulation impaired synaptic plasticity through STAT3 inactivation induced suppression of NMDARs expression, revealing a novel mechanism for hTau-associated synapse and memory deficits.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Gong-Ping Liu
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, JS 226001, China
| |
Collapse
|
59
|
Ni J, Wu Z. Inflammation Spreading: Negative Spiral Linking Systemic Inflammatory Disorders and Alzheimer's Disease. Front Cell Neurosci 2021; 15:638686. [PMID: 33716675 PMCID: PMC7947253 DOI: 10.3389/fncel.2021.638686] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 02/03/2021] [Indexed: 12/15/2022] Open
Abstract
As a physiological response to injury in the internal body organs, inflammation is responsible for removing dangerous stimuli and initiating healing. However, persistent and exaggerative chronic inflammation causes undesirable negative effects in the organs. Inflammation occurring in the brain and spinal cord is known as neuroinflammation, with microglia acting as the central cellular player. There is increasing evidence suggesting that chronic neuroinflammation is the most relevant pathological feature of Alzheimer’s disease (AD), regulating other pathological features, such as the accumulation of amyloid-β (Aβ) and hyperphosphorylation of Tau. Systemic inflammatory signals caused by systemic disorders are known to strongly influence neuroinflammation as a consequence of microglial activation, inflammatory mediator production, and the recruitment of peripheral immune cells to the brain, resulting in neuronal dysfunction. However, the neuroinflammation-accelerated neuronal dysfunction in AD also influences the functions of peripheral organs. In the present review, we highlight the link between systemic inflammatory disorders and AD, with inflammation serving as the common explosion. We discuss the molecular mechanisms that govern the crosstalk between systemic inflammation and neuroinflammation. In our view, inflammation spreading indicates a negative spiral between systemic diseases and AD. Therefore, “dampening inflammation” through the inhibition of cathepsin (Cat)B or CatS may be a novel therapeutic approach for delaying the onset of and enacting early intervention for AD.
Collapse
Affiliation(s)
- Junjun Ni
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Zhou Wu
- Department of Aging Science and Pharmacology, Faculty of Dental Science, Kyushu University, Fukuoka, Japan.,OBT Research Center, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| |
Collapse
|
60
|
Reischer G, Heinke B, Sandkühler J. Interferon-γ facilitates the synaptic transmission between primary afferent C-fibres and lamina I neurons in the rat spinal dorsal horn via microglia activation. Mol Pain 2021; 16:1744806920917249. [PMID: 32264753 PMCID: PMC7144669 DOI: 10.1177/1744806920917249] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Recent studies have demonstrated an important role of the pro-inflammatory cytokine interferon-γ in neuropathic pain. Interferon-γ is upregulated in the lumbar spinal cord of nerve-injured rodents and intrathecal injection of interferon-γ has been shown to induce neuropathic pain-like behaviours in naive rodents. A potential mechanism in the pathogenesis of neuropathic pain is a long-lasting amplification of nociceptive synaptic transmission in lamina I of the spinal dorsal horn. Here, we tested the effects of interferon-γ on the properties of the first synapse in nociceptive pathways in the superficial spinal dorsal horn. We performed whole-cell patch-clamp recordings in lamina I neurons in a spinal cord slice preparation with dorsal roots attached from young rats. We determined the effects of acute (at least 25 min) or longer lasting (4–8 h) treatment of the transversal slices with recombinant rat interferon-γ on spontaneous excitatory postsynaptic currents or on monosynaptic Aδ- and C-fibre-evoked excitatory postsynaptic currents, respectively. Prolonged treatment with interferon-γ facilitated monosynaptic C-fibre-evoked excitatory postsynaptic currents and this effect could be blocked by co-application of minocycline an inhibitor of microglial activation. In contrast, Aδ-fibre-evoked excitatory postsynaptic currents were not affected by the prolonged interferon-γ treatment. Acute interferon-γ application in the bathing solution did not change strength of monosynaptic Aδ- or C-fibre synapses in lamina I. However, the rate, but not the amplitude, of spontaneous excitatory postsynaptic currents recorded in lamina I neurons was decreased. This effect could not be blocked by the application of minocycline. Long-lasting treatment of rat spinal cord slices with interferon-γ induced an input specific facilitation of synaptic strength in spinal nociceptive pathways. Enhanced transmission between C-fibres and spinal lamina I neurons was mediated by the activation of microglial cells. We showed that the pro-inflammatory cytokine interferon-γ modifies the processing of information at the first synaptic relay station in nociceptive pathways.
Collapse
Affiliation(s)
- Gerda Reischer
- Department of Neurophysiology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Bernhard Heinke
- Department of Neurophysiology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Jürgen Sandkühler
- Department of Neurophysiology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
61
|
Han JK, Kwon SH, Kim YG, Choi J, Kim JI, Lee YS, Ye SK, Kim SJ. Ablation of STAT3 in Purkinje cells reorganizes cerebellar synaptic plasticity in long-term fear memory network. eLife 2021; 10:e63291. [PMID: 33459594 PMCID: PMC7813544 DOI: 10.7554/elife.63291] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 12/31/2020] [Indexed: 12/15/2022] Open
Abstract
Emotional memory processing engages a large neuronal network of brain regions including the cerebellum. However, the molecular and cellular mechanisms of the cerebellar cortex modulating the fear memory network are unclear. Here, we illustrate that synaptic signaling in cerebellar Purkinje cells (PCs) via STAT3 regulates long-term fear memory. Transcriptome analyses revealed that PC-specific STAT3 knockout (STAT3PKO) results in transcriptional changes that lead to an increase in the expression of glutamate receptors. The amplitude of AMPA receptor-mediated excitatory postsynaptic currents at parallel fiber (PF) to PC synapses was larger in STAT3PKO mice than in wild-type (WT) littermates. Fear conditioning induced long-term depression of PF-PC synapses in STAT3PKO mice while the same manipulation induced long-term potentiation in WT littermates. STAT3PKO mice showed an aberrantly enhanced long-term fear memory. Neuronal activity in fear-related regions increased in fear-conditioned STAT3PKO mice. Our data suggest that STAT3-dependent molecular regulation in PCs is indispensable for proper expression of fear memory.
Collapse
Affiliation(s)
- Jeong-Kyu Han
- Department of Physiology, Seoul National University College of MedicineSeoulRepublic of Korea
- Department of Brain and Cognitive Sciences, Seoul National University Graduate SchoolSeoulRepublic of Korea
- Memory Network Medical Research Center, Seoul National University College of MedicineSeoulRepublic of Korea
- Neuroscience Research Institute, Seoul National University College of MedicineSeoulRepublic of Korea
| | - Sun-Ho Kwon
- Memory Network Medical Research Center, Seoul National University College of MedicineSeoulRepublic of Korea
- Department of Pharmacology, Seoul National University College of MedicineSeoulRepublic of Korea
- Department of Biomedical Sciences, Seoul National University College of MedicineSeoulRepublic of Korea
| | - Yong Gyu Kim
- Department of Physiology, Seoul National University College of MedicineSeoulRepublic of Korea
- Department of Biomedical Sciences, Seoul National University College of MedicineSeoulRepublic of Korea
| | - Jaeyong Choi
- Department of Biomedical Sciences, Seoul National University College of MedicineSeoulRepublic of Korea
- Department of Biochemistry and Molecular Biology, Seoul National University College of MedicineSeoulRepublic of Korea
| | - Jong-Il Kim
- Department of Biomedical Sciences, Seoul National University College of MedicineSeoulRepublic of Korea
- Department of Biochemistry and Molecular Biology, Seoul National University College of MedicineSeoulRepublic of Korea
| | - Yong-Seok Lee
- Department of Physiology, Seoul National University College of MedicineSeoulRepublic of Korea
- Neuroscience Research Institute, Seoul National University College of MedicineSeoulRepublic of Korea
- Department of Biomedical Sciences, Seoul National University College of MedicineSeoulRepublic of Korea
| | - Sang-Kyu Ye
- Department of Physiology, Seoul National University College of MedicineSeoulRepublic of Korea
- Department of Pharmacology, Seoul National University College of MedicineSeoulRepublic of Korea
- Department of Biomedical Sciences, Seoul National University College of MedicineSeoulRepublic of Korea
| | - Sang Jeong Kim
- Department of Brain and Cognitive Sciences, Seoul National University Graduate SchoolSeoulRepublic of Korea
- Memory Network Medical Research Center, Seoul National University College of MedicineSeoulRepublic of Korea
- Neuroscience Research Institute, Seoul National University College of MedicineSeoulRepublic of Korea
- Department of Biomedical Sciences, Seoul National University College of MedicineSeoulRepublic of Korea
| |
Collapse
|
62
|
Espírito-Santo SA, Nunes-Tavares N, Mendonça HR, Serfaty CA, Sholl-Franco A, Campello-Costa P. Intravitreal Interleukin-2 modifies retinal excitatory circuits and retinocollicular innervation. Exp Eye Res 2021; 204:108442. [PMID: 33460624 DOI: 10.1016/j.exer.2021.108442] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 12/14/2020] [Accepted: 01/08/2021] [Indexed: 10/22/2022]
Abstract
Interleukin-2 is a classical immune cytokine whose neural functions have received little attention. Its levels have been found to be increased in some neuropathologies, such as Alzheimer's disease, multiple sclerosis and uveitis. Mechanistically, it has been demonstrated the role of IL-2 in regulating glutamate and acetylcholine transmission, thus being relevant for CNS physiology. In fact, our previous work showed that an acute intravitreal IL-2 injection during retinotectal development promoted contralateral eye axonal plasticity in the superior colliculus, but the involved mechanisms were not explored. So, our present study aimed to investigate the effect of increased intravitreal IL-2 levels on the retinal glutamatergic and cholinergic signalling required for retinotectal normal development. We showed through HRP neuronal tracing that intravitreal IL-2 also induces ipsilateral eye axonal sprouting. Protein level and/or immunolocalization analysis in the retina confirmed IL-2 pathway activation by increased expression of phospho-STAT-3, coupled to transient (24h) reduced levels of Egr1, PSD-95 and nicotinic acetylcholine receptor β2 subunit, suggesting reduced neural activity and synaptic sites. Also, AChE activity and GluN2B and GluA2 contents were reduced within 96h after IL-2 treatment. Therefore, IL-2-induced retinotectal plasticity might be driven by changes in cholinergic and glutamatergic pathways of the retina.
Collapse
Affiliation(s)
- S A Espírito-Santo
- Instituto de Biologia, Programa de Pós-Graduação Em Neurociências, Universidade Federal Fluminense, Niterói, Brazil; Instituto de Biofísica, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil; Universidade Do Estado de Minas Gerais, Departamento de Ciências Biológicas, Minas Gerais, Brazil
| | - N Nunes-Tavares
- Instituto de Biofísica, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - H R Mendonça
- Instituto de Biologia, Programa de Pós-Graduação Em Neurociências, Universidade Federal Fluminense, Niterói, Brazil; Laboratório Integrado de Morfologia, Instituto de Biodiversidade e Sustentabilidade NUPEM, Universidade Federal Do Rio de Janeiro, Campus Macaé, Brazil
| | - C A Serfaty
- Instituto de Biologia, Programa de Pós-Graduação Em Neurociências, Universidade Federal Fluminense, Niterói, Brazil
| | - A Sholl-Franco
- Instituto de Biofísica, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - P Campello-Costa
- Instituto de Biologia, Programa de Pós-Graduação Em Neurociências, Universidade Federal Fluminense, Niterói, Brazil.
| |
Collapse
|
63
|
Hamilton K, Harvey J. The Neuronal Actions of Leptin and the Implications for Treating Alzheimer's Disease. Pharmaceuticals (Basel) 2021; 14:ph14010052. [PMID: 33440796 PMCID: PMC7827292 DOI: 10.3390/ph14010052] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/06/2021] [Accepted: 01/07/2021] [Indexed: 12/13/2022] Open
Abstract
It is widely accepted that the endocrine hormone leptin controls food intake and energy homeostasis via activation of leptin receptors expressed on hypothalamic arcuate neurons. The hippocampal formation also displays raised levels of leptin receptor expression and accumulating evidence indicates that leptin has a significant impact on hippocampal synaptic function. Thus, cellular and behavioural studies support a cognitive enhancing role for leptin as excitatory synaptic transmission, synaptic plasticity and glutamate receptor trafficking at hippocampal Schaffer collateral (SC)-CA1 synapses are regulated by leptin, and treatment with leptin enhances performance in hippocampus-dependent memory tasks. Recent studies indicate that hippocampal temporoammonic (TA)-CA1 synapses are also a key target for leptin. The ability of leptin to regulate TA-CA1 synapses has important functional consequences as TA-CA1 synapses are implicated in spatial and episodic memory processes. Moreover, degeneration is initiated in the TA pathway at very early stages of Alzheimer's disease, and recent clinical evidence has revealed links between plasma leptin levels and the incidence of Alzheimer's disease (AD). Additionally, accumulating evidence indicates that leptin has neuroprotective actions in various AD models, whereas dysfunctions in the leptin system accelerate AD pathogenesis. Here, we review the data implicating the leptin system as a potential novel target for AD, and the evidence that boosting the hippocampal actions of leptin may be beneficial.
Collapse
|
64
|
Yu L, Wen G, Zhu S, Hu X, Huang C, Yang Y. Abnormal phosphorylation of tau protein and neuroinflammation induced by laparotomy in an animal model of postoperative delirium. Exp Brain Res 2021; 239:867-880. [PMID: 33409674 DOI: 10.1007/s00221-020-06007-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 12/05/2020] [Indexed: 10/22/2022]
Abstract
Postoperative delirium (POD) is an acute neuropsychological disturbance after surgery, whose prevalence is related with advancing age. Neuroinflammation and abnormal tau phosphorylation that commonly presenting in Alzheimer's disease (AD) may contribute to the progression and duration of POD. To study the acute influence of surgery on cognitive function, wild type male C57BL/6 N mice were randomly divided into three groups: Control (CON), Laparotomy at 4 h and 24 h (LAP-4 h, LAP-24 h), then subjected to laparotomy under sevoflurane anaesthesia. The cognitive performance, peripheral and central inflammatory responses and tau phosphorylation levels were evaluated at 4 h and 24 h postoperatively. When LAP4-hrs displayed anxiety behaviors with high mRNA levels of inflammatory cytokines, such as interleukin-1β (IL-1β), IL-6, IL-8, TNF-α and MCP-1 in the liver, and IL-8 in the hippocampus, results at 24 h were different. In the liver, only IL-10 protein was obviously elevated, but in the hippocampus, both pro- and anti-inflammatory cytokines were significantly decreased whilst the elimination of anxiety. The activity of major related kinases and phosphatases was remarkably changed which may contribute to the dephosphorylated tau protein. With tremendous neuropathological changes and significant numbers of activated microglias and astrocytes observed in the sub-regions of hippocampus, the memory impairment existed at both 4 h and 24 h. Since the association of dephosphorylated tau with POD, these findings may supply novel implications for the understanding of tauopathies and as a theoretical basis for preventions from the postoperative cognitive dysfunction (POCD).
Collapse
Affiliation(s)
- Le Yu
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-Inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230001, People's Republic of China.,Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230001, People's Republic of China.,Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Hefei, Anhui, People's Republic of China
| | - Guanghua Wen
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-Inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230001, People's Republic of China
| | - Shoufeng Zhu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230001, People's Republic of China.,Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Hefei, Anhui, People's Republic of China
| | - Xianwen Hu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230001, People's Republic of China.,Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Hefei, Anhui, People's Republic of China
| | - Chunxia Huang
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230001, People's Republic of China. .,Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Hefei, Anhui, People's Republic of China.
| | - Yan Yang
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-Inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230001, People's Republic of China.
| |
Collapse
|
65
|
Zhang M, Wang Y, Zhu G, Sun C, Wang J. Hepatoprotective effect and possible mechanism of phytoestrogen calycosin on carbon tetrachloride-induced liver fibrosis in mice. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2021; 394:189-204. [PMID: 32474674 DOI: 10.1007/s00210-020-01891-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 04/28/2020] [Indexed: 01/02/2023]
Abstract
The study was to explore the hepatoprotective effect and possible mechanism of calycosin on carbon tetrachloride (CCl4)-induced liver fibrosis in mice. Hepatic fibrosis was induced by intraperitoneal injection of CCl4 in C57BL/6 male mice. Serum alanine aminotransferase (ALT) and aspartate transaminase (AST) activity, superoxide dismutase (SOD) activity, and hydroxyproline (Hyp) and malondialdehyde (MDA) levels were determined by biochemical assays. Liver histopathology was assessed by H&E and Masson trichrome staining. The mRNA expressions of α-smooth muscle actin (α-SMA), collagen-I (Col-I), Janus kinase 2 (JAK2) and signal transducer and activator of transcription 3 (STAT3) were determined using qRT-PCR. The protein levels of α-SMA, Col-I, estrogen receptor α (ERα), estrogen receptor β (ERβ), tissue inhibitor of metalloproteinase-1 (TIMP-1), matrix metalloproteinase-1 (MMP-1), JAK2, phospho-JAK2 (p-JAK2), STAT3, and phospho-STAT3 (p-STAT3) were detected by Western blotting. The levels of α-SMA and ERβ were measured by immunohistochemistry. Calycosin significantly reduced liver index, MDA level, and ALT and AST activity and increased SOD activity. The α-SMA, Col-I, and Hyp of the calycosin group were significantly lower than those of the model group. Calycosin increased MMP-1 and inhibited TIMP-1 expression resulting in the improvement of MMP-1/TIMP-1 ratio. Importantly, calycosin improved ERβ protein expression, JAK2 and STAT3 mRNA expressions, p-JAK2/JAK2, and p-STAT3/STAT3 relative protein expressions. However, ERα, JAK2, and STAT3 protein expressions were relatively unchanged. Calycosin significantly inhibits liver fibrosis in mice, and its mechanism may involve the following: calycosin inhibits oxidative stress; calycosin inhibits collagen synthesis and balances MMP-1/TIMP-1 system; calycosin increases ERβ expression and activates JAK2-STAT3 pathway.
Collapse
Affiliation(s)
- Mengmeng Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, Anhui Province, China
| | - Yaxin Wang
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, Anhui Province, China
| | - Guannan Zhu
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, Anhui Province, China
| | - Cheng Sun
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, Anhui Province, China
| | - Jiajia Wang
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, Anhui Province, China.
| |
Collapse
|
66
|
STAT3 in the dorsal raphe gates behavioural reactivity and regulates gene networks associated with psychopathology. Mol Psychiatry 2021; 26:2886-2899. [PMID: 33046834 PMCID: PMC8505245 DOI: 10.1038/s41380-020-00904-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 09/30/2020] [Accepted: 10/01/2020] [Indexed: 01/02/2023]
Abstract
The signal transducer and activator of transcription 3 (STAT3) signalling pathway is activated through phosphorylation by Janus kinases in response to a diverse set of immunogenic and non-immunogenic triggers. Several distinct lines of evidence propose an intricate involvement of STAT3 in neural function relevant to behaviour in health and disease. However, in part due to the pleiotropic effects resulting from its DNA binding activity and the consequent regulation of expression of a variety of genes with context-dependent cellular consequences, the precise nature of STAT3 involvement in the neural mechanisms underlying psychopathology remains incompletely understood. Here, we focused on the midbrain serotonergic system, a central hub for the regulation of emotions, to examine the relevance of STAT3 signalling for emotional behaviour in mice by selectively knocking down raphe STAT3 expression using germline genetic (STAT3 KO) and viral-mediated approaches. Mice lacking serotonergic STAT3 presented with reduced negative behavioural reactivity and a blunted response to the sensitising effects of amphetamine, alongside alterations in midbrain neuronal firing activity of serotonergic neurons and transcriptional control of gene networks relevant for neuropsychiatric disorders. Viral knockdown of dorsal raphe (DR) STAT3 phenocopied the behavioural alterations of STAT3 KO mice, excluding a developmentally determined effect and suggesting that disruption of STAT3 signalling in the DR of adult mice is sufficient for the manifestation of behavioural traits relevant to psychopathology. Collectively, these results suggest DR STAT3 as a molecular gate for the control of behavioural reactivity, constituting a mechanistic link between the upstream activators of STAT3, serotonergic neurotransmission and psychopathology.
Collapse
|
67
|
Zamora-Moratalla A, Martín ED. Prolactin enhances hippocampal synaptic plasticity in female mice of reproductive age. Hippocampus 2020; 31:281-293. [PMID: 33285014 PMCID: PMC7983975 DOI: 10.1002/hipo.23288] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 11/17/2020] [Accepted: 11/20/2020] [Indexed: 01/27/2023]
Abstract
Dynamic signaling between the endocrine system (ES) and the nervous system (NS) is essential for brain and body homeostasis. In particular, reciprocal interaction occurs during pregnancy and motherhood that may involve changes in some brain plasticity processes. Prolactin (PRL), a hormone with pleiotropic effects on the NS, promotes maternal behavior and has been linked to modifications in brain circuits during motherhood; however, it is unclear whether PRL may regulate synaptic plasticity. Therefore, the main aim of the present work was to determine the cellular and molecular mechanisms triggered by PRL that regulate synaptic plasticity in the hippocampus. By analyzing extracellular recordings in CA3‐CA1 synapses of hippocampal slices, we report that PRL modifies short and long‐term synaptic plasticity in female mice of reproductive age, but not in sexually immature females or adult males. This effect is carried out through mechanisms that include participation of GABAA receptors and activation of the JAK2‐mediated signaling pathway. These findings show for the first time how PRL enhances the synaptic strength in hippocampal circuits and that this effect is sexually dimorphic, which would influence complex brain processes in physiological conditions like pregnancy and lactation.
Collapse
Affiliation(s)
- Alfonsa Zamora-Moratalla
- Laboratory of Neurophysiology and Synaptic Plasticity, Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Eduardo D Martín
- Laboratory of Neurophysiology and Synaptic Plasticity, Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| |
Collapse
|
68
|
Toral-Rios D, Patiño-López G, Gómez-Lira G, Gutiérrez R, Becerril-Pérez F, Rosales-Córdova A, León-Contreras JC, Hernández-Pando R, León-Rivera I, Soto-Cruz I, Florán-Garduño B, Campos-Peña V. Activation of STAT3 Regulates Reactive Astrogliosis and Neuronal Death Induced by AβO Neurotoxicity. Int J Mol Sci 2020; 21:ijms21207458. [PMID: 33050466 PMCID: PMC7590075 DOI: 10.3390/ijms21207458] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/23/2020] [Accepted: 09/25/2020] [Indexed: 01/03/2023] Open
Abstract
Amyloid-beta oligomers (AβO) have been proposed as the most potent neurotoxic and inflammation inducers in Alzheimer’s disease (AD). AβO contribute to AD pathogenesis by impairing the production of several cytokines and inflammation-related signaling pathways, such as the Janus kinases/signal transducer of transcription factor-3 (JAK/STAT3) pathway. STAT3 modulates glial activation, indirectly regulates Aβ deposition, and induces cognitive decline in AD transgenic models. However, in vivo studies using an AβO microinjection rat model have not yet explored STAT3 role. The main purpose of this study was to elucidate if a single microinjection of AβO could promote an increased expression of STAT3 in glial cells favoring neuroinflammation and neurodegeneration. We designed a model of intrahippocampal microinjection and assessed glial activation, cytokines production, STAT3 expression, and neurodegeneration in time. Our results showed robust expression of STAT3 in glial cells (mainly in astrocytes) and neurons, correlating with neuronal death in response to AβO administration. A STAT3 inhibition assay conducted in rat primary hippocampal cultures, suggested that the induction of the transcription factor by AβO in astrocytes leads them to an activation state that may favor neuronal death. Notwithstanding, pharmacological inhibition of the JAK2/STAT3 pathway should be focused on astrocytes because it is also essential in neurons survival. Overall, these findings strongly suggest the participation of STAT3 in the development of neurodegeneration.
Collapse
Affiliation(s)
- Danira Toral-Rios
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de Mexico 07360, Mexico; (D.T.-R.); (B.F.-G.)
| | - Genaro Patiño-López
- Laboratorio de Investigación en Inmunología y Proteómica, Hospital Infantil de México Federico Gómez, Ciudad de Mexico 06720, Mexico;
| | - Gisela Gómez-Lira
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de Mexico 14330, Mexico; (G.G.-L.); (R.G.)
| | - Rafael Gutiérrez
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de Mexico 14330, Mexico; (G.G.-L.); (R.G.)
| | - Fernando Becerril-Pérez
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Campus-Vienna-BioCenter 1, 1030 Vienna, Austria;
| | - Aldebarán Rosales-Córdova
- Departamento de Administración, Facultad de Economía y Negocios, Universidad Anáhuac de México, Huixquilucan 52786, Mexico;
| | - Juan Carlos León-Contreras
- Departamento de Patología, Sección Patología Experimental, Instituto Nacional de Ciencias Médicas y Nutrición, Salvador Zubirán, Ciudad de Mexico 14080, Mexico; (J.C.L.-C.); (R.H.-P.)
| | - Rogelio Hernández-Pando
- Departamento de Patología, Sección Patología Experimental, Instituto Nacional de Ciencias Médicas y Nutrición, Salvador Zubirán, Ciudad de Mexico 14080, Mexico; (J.C.L.-C.); (R.H.-P.)
| | - Ismael León-Rivera
- Centro de Investigaciones Químicas, IICBA, Universidad Autónoma del Estado de Morelos, Cuernavaca Morelos 62210, Mexico;
| | - Isabel Soto-Cruz
- Laboratorio de Oncología Molecular, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Ciudad de Mexico 09230, Mexico;
| | - Benjamín Florán-Garduño
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de Mexico 07360, Mexico; (D.T.-R.); (B.F.-G.)
| | - Victoria Campos-Peña
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Ciudad de Mexico 14269, Mexico
- Correspondence: ; Tel.: +555-6063-822 (ext. 2010)
| |
Collapse
|
69
|
The protective effects of prolactin on brain injury. Life Sci 2020; 263:118547. [PMID: 33038380 DOI: 10.1016/j.lfs.2020.118547] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/23/2020] [Accepted: 09/28/2020] [Indexed: 11/22/2022]
Abstract
AIMS Brain injuries based on their causes are divided into two categories, TBI and NTBI. TBI is caused by damages such as head injury, but non-physical injury causes NTBI. Prolactin is one of the blood factors that increase during brain injury. It has been assumed to play a regenerative role in post-injury recovery. MATERIALS AND METHODS In this review, various valid papers from electronic sources (including Web of Science, Scopus, PubMed, SID, Google Scholar, and ISI databases) used, which in them the protective effect of prolactin on brain injury investigated. KEY FINDINGS Inflammation following brain injury with the production of pro-inflammatory cytokines in the affected area can even lead to excitotoxicity and cell death in the damaged area. Medical brain damage treatments are long-term, and can have several side effects. Therefore, it is better to consider medication treatments that have fewer side effects and greater efficacy. Research suggests that prolactin has numerous regenerative effects on brain injury, and prevents cell death. Prolactin is one of the hormones produced in the body; therefore it has fewer side effects and may be more effective because it increases during brain injury. SIGNIFICANCE Prolactin can be used peripherally and centrally, and exerts its neuro regenerative effects against further damage post-TBI and NTBI.
Collapse
|
70
|
Lago SG, Tomasik J, van Rees GF, Ramsey JM, Haenisch F, Cooper JD, Broek JA, Suarez-Pinilla P, Ruland T, Auyeug B, Mikova O, Kabacs N, Arolt V, Baron-Cohen S, Crespo-Facorro B, Bahn S. Exploring the neuropsychiatric spectrum using high-content functional analysis of single-cell signaling networks. Mol Psychiatry 2020; 25:2355-2372. [PMID: 30038233 DOI: 10.1038/s41380-018-0123-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 05/04/2018] [Accepted: 05/25/2018] [Indexed: 12/26/2022]
Abstract
Neuropsychiatric disorders overlap in symptoms and share genetic risk factors, challenging their current classification into distinct diagnostic categories. Novel cross-disorder approaches are needed to improve our understanding of the heterogeneous nature of neuropsychiatric diseases and overcome existing bottlenecks in their diagnosis and treatment. Here we employ high-content multi-parameter phospho-specific flow cytometry, fluorescent cell barcoding and automated sample preparation to characterize ex vivo signaling network responses (n = 1764) measured at the single-cell level in B and T lymphocytes across patients diagnosed with four major neuropsychiatric disorders: autism spectrum condition (ASC), bipolar disorder (BD), major depressive disorder (MDD), and schizophrenia (SCZ; n = 25 each), alongside matched healthy controls (n = 100). We identified 25 nodes (individual cell subtype-epitope-ligand combinations) significantly altered relative to the control group, with variable overlap between different neuropsychiatric diseases and heterogeneously expressed at the level of each individual patient. Reconstruction of the diagnostic categories from the altered nodes revealed an overlapping neuropsychiatric spectrum extending from MDD on one end, through BD and SCZ, to ASC on the other end. Network analysis showed that although the pathway structure of the epitopes was broadly preserved across the clinical groups, there were multiple discrete alterations in network connectivity, such as disconnections within the antigen/integrin receptor pathway and increased negative regulation within the Akt1 pathway in CD4+ T cells from ASC and SCZ patients, in addition to increased correlation of Stat1 (pY701) and Stat5 (pY694) responses in B cells from BD and MDD patients. Our results support the "dimensional" approach to neuropsychiatric disease classification and suggest potential novel drug targets along the neuropsychiatric spectrum.
Collapse
Affiliation(s)
- Santiago G Lago
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Jakub Tomasik
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Geertje F van Rees
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Jordan M Ramsey
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Frieder Haenisch
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Jason D Cooper
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Jantine A Broek
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Paula Suarez-Pinilla
- Department of Psychiatry, Marqués de Valdecilla University Hospital, IDIVAL, School of Medicine, University of Cantabria, Santander, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Santander, Spain
| | - Tillmann Ruland
- Department of Psychiatry and Psychotherapy, University of Münster, Münster, Germany
| | - Bonnie Auyeug
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, UK.,Psychology Department, Edinburgh University, Scotland, UK
| | - Olya Mikova
- Foundation Biological Psychiatry, Sofia, Bulgaria
| | - Nikolett Kabacs
- Cambridgeshire and Peterborough NHS Foundation Trust, Cambridge, UK
| | - Volker Arolt
- Department of Psychiatry and Psychotherapy, University of Münster, Münster, Germany
| | - Simon Baron-Cohen
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, UK.,CLASS Clinic, Cambridgeshire and Peterborough NHS Foundation Trust, Cambridge, UK
| | - Benedicto Crespo-Facorro
- Department of Psychiatry, Marqués de Valdecilla University Hospital, IDIVAL, School of Medicine, University of Cantabria, Santander, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Santander, Spain
| | - Sabine Bahn
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
71
|
Activation of tyrosine phosphatase PTP1B in pyramidal neurons impairs endocannabinoid signaling by tyrosine receptor kinase trkB and causes schizophrenia-like behaviors in mice. Neuropsychopharmacology 2020; 45:1884-1895. [PMID: 32610340 PMCID: PMC7608138 DOI: 10.1038/s41386-020-0755-3] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/19/2020] [Accepted: 06/24/2020] [Indexed: 12/12/2022]
Abstract
Schizophrenia is a debilitating disorder affecting young adults displaying symptoms of cognitive impairment, anxiety, and early social isolation prior to episodes of auditory hallucinations. Cannabis use has been tied to schizophrenia-like symptoms, indicating that dysregulated endogenous cannabinoid signaling may be causally linked to schizophrenia. Previously, we reported that glutamatergic neuron-selective ablation of Lmo4, an endogenous inhibitor of the tyrosine phosphatase PTP1B, impairs endocannabinoid (eCB) production from the metabotropic glutamate receptor mGluR5. These Lmo4-deficient mice display anxiety-like behaviors that are alleviated by local shRNA knockdown or pharmacological inhibition of PTP1B that restores mGluR5-dependent eCB production in the amygdala. Here, we report that these Lmo4-deficient mice also display schizophrenia-like behaviors: impaired working memory assessed in the Y maze and defective sensory gating by prepulse inhibition of the acoustic startle response. Modulation of inhibitory inputs onto layer 2/3 pyramidal neurons of the prefrontal cortex relies on eCB signaling from the brain-derived neurotrophic factor receptor trkB, rather than mGluR5, and this mechanism was defective in Lmo4-deficient mice. Genetic ablation of PTP1B in the glutamatergic neurons lacking Lmo4 restored tyrosine phosphorylation of trkB, trkB-mediated eCB signaling, and ameliorated schizophrenia-like behaviors. Pharmacological inhibition of PTP1B with trodusquemine also restored trkB phosphorylation and improved schizophrenia-like behaviors by restoring eCB signaling, since the CB1 receptor antagonist 1-(2,4-dichlorophenyl)-5-(4-iodophenyl)-4-methyl-N-1-piperidinyl-1H-pyrazole-3-carboxamide blocked this effect. Thus, activation of PTP1B in pyramidal neurons contributes to schizophrenia-like behaviors in Lmo4-deficient mice and genetic or pharmacological intervention targeting PTP1B ameliorates schizophrenia-related deficits.
Collapse
|
72
|
Wadsworth PA, Singh AK, Nguyen N, Dvorak NM, Tapia CM, Russell WK, Stephan C, Laezza F. JAK2 regulates Nav1.6 channel function via FGF14 Y158 phosphorylation. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2020; 1867:118786. [PMID: 32599005 PMCID: PMC7984254 DOI: 10.1016/j.bbamcr.2020.118786] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 06/18/2020] [Accepted: 06/20/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Protein interactions between voltage-gated sodium (Nav) channels and accessory proteins play an essential role in neuronal firing and plasticity. However, a surprisingly limited number of kinases have been identified as regulators of these molecular complexes. We hypothesized that numerous as-of-yet unidentified kinases indirectly regulate the Nav channel via modulation of the intracellular fibroblast growth factor 14 (FGF14), an accessory protein with numerous unexplored phosphomotifs and required for channel function in neurons. METHODS Here we present results from an in-cell high-throughput screening (HTS) against the FGF14: Nav1.6 complex using >3000 diverse compounds targeting an extensive range of signaling pathways. Regulation by top kinase targets was then explored using in vitro phosphorylation, biophysics, mass-spectrometry and patch-clamp electrophysiology. RESULTS Compounds targeting Janus kinase 2 (JAK2) were over-represented among HTS hits. Phosphomotif scans supported by mass spectrometry revealed FGF14Y158, a site previously shown to mediate both FGF14 homodimerization and interactions with Nav1.6, as a JAK2 phosphorylation site. Following inhibition of JAK2, FGF14 homodimerization increased in a manner directly inverse to FGF14:Nav1.6 complex formation, but not in the presence of the FGF14Y158A mutant. Patch-clamp electrophysiology revealed that through Y158, JAK2 controls FGF14-dependent modulation of Nav1.6 channels. In hippocampal CA1 pyramidal neurons, the JAK2 inhibitor Fedratinib reduced firing by a mechanism that is dependent upon expression of FGF14. CONCLUSIONS These studies point toward a novel mechanism by which levels of JAK2 in neurons could directly influence firing and plasticity by controlling the FGF14 dimerization equilibrium, and thereby the availability of monomeric species for interaction with Nav1.6.
Collapse
Affiliation(s)
- Paul A Wadsworth
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, USA; Department of Pharmacology & Toxicology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Aditya K Singh
- Department of Pharmacology & Toxicology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Nghi Nguyen
- HTS Screening Core, Center for Translational Cancer Research, Texas A&M Health Science Center: Institute of Biosciences and Technology, Houston, TX, USA
| | - Nolan M Dvorak
- Department of Pharmacology & Toxicology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Cynthia M Tapia
- Department of Pharmacology & Toxicology, The University of Texas Medical Branch, Galveston, TX, USA
| | - William K Russell
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Clifford Stephan
- HTS Screening Core, Center for Translational Cancer Research, Texas A&M Health Science Center: Institute of Biosciences and Technology, Houston, TX, USA
| | - Fernanda Laezza
- Department of Pharmacology & Toxicology, The University of Texas Medical Branch, Galveston, TX, USA; Center for Addiction Research, The University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
73
|
Decreased motor impulsivity following chronic lithium treatment in male rats is associated with reduced levels of pro-inflammatory cytokines in the orbitofrontal cortex. Brain Behav Immun 2020; 89:339-349. [PMID: 32688024 DOI: 10.1016/j.bbi.2020.07.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/30/2020] [Accepted: 07/13/2020] [Indexed: 02/06/2023] Open
Abstract
Lithium's efficacy in reducing both symptom severity in bipolar disorder (BD) and suicide risk across clinical populations may reflect its ability to reduce impulsivity. Changes in immune markers are associated with BD and suicidality yet their exact role in symptom expression remains unknown. Evidence also suggests that lithium may decrease levels of pro-inflammatory cytokines in the periphery and central nervous system, and that such changes are related to its therapeutic efficacy. However, issues of cause and effect are hard to infer from clinical data alone. Here, we investigated the effects of chronic dietary lithium treatment on rats' performance of the 5-Choice Serial Reaction Time Task (5CSRTT), a well-validated operant behavioural task measuring aspects of impulsivity, attention and motivation. Male Long-Evans rats received a diet supplemented with 0.3% LiCl (n = 13), or the equivalent control diet (n = 16), during behavioural testing. Blood and brain tissue samples were assayed for a wide range of cytokines once any changes in impulsivity became significant. After 12 weeks, chronic lithium treatment reduced levels of motor impulsivity, as indexed by premature responses in the 5CSRTT; measures of sustained attention and motivation were unaffected. Plasma levels of IL-1β, IL-10 and RANTES (CCL-5) were reduced in lithium-treated rats at this time point. IL-1β, IL-6 and RANTES were also reduced selectively within the orbitofrontal cortex of lithium-treated rats, whereas cytokine levels in the medial prefrontal cortex and nucleus accumbens were comparable with control subjects. These results are consistent with the hypothesis that lithium may improve impulse control deficits in clinical populations by minimising the effects of pro-inflammatory signalling on neuronal activity, particularly within the orbitofrontal cortex.
Collapse
|
74
|
Worthen RJ, Garzon Zighelboim SS, Torres Jaramillo CS, Beurel E. Anti-inflammatory IL-10 administration rescues depression-associated learning and memory deficits in mice. J Neuroinflammation 2020; 17:246. [PMID: 32828124 PMCID: PMC7443292 DOI: 10.1186/s12974-020-01922-1] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 08/10/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Major depressive disorder is a widespread mood disorder. One of the most debilitating symptoms patients often experience is cognitive impairment. Recent findings suggest that inflammation is associated with depression and impaired cognition. Pro-inflammatory cytokines are elevated in the blood of depressed patients and impair learning and memory processes, suggesting that an anti-inflammatory approach might be beneficial for both depression and cognition. METHODS We subjected mice to the learned helplessness paradigm and evaluated novel object recognition and spatial memory. Mice were treated with IL-10 intranasally or/and microglia cells were depleted using PLX5622. Statistical differences were tested using ANOVA or t tests. RESULTS We first established a mouse model of depression in which learning and memory are impaired. We found that learned helplessness (LH) impairs novel object recognition (NOR) and spatial working memory. LH mice also exhibit reduced hippocampal dendritic spine density and increased microglial activation compared to non-shocked (NS) mice or mice that were subjected to the learned helpless paradigm but did not exhibit learned helplessness (non-learned helpless or NLH). These effects are mediated by microglia, as treatment with PLX5622, which depletes microglia, restores learning and memory and hippocampal dendritic spine density in LH mice. However, PLX5622 also impairs learning and memory and reduces hippocampal dendritic spine density in NLH mice, suggesting that microglia in NLH mice produce molecules that promote learning and memory. We found that microglial interleukin (IL)-10 levels are reduced in LH mice, and IL-10 administration is sufficient to restore NOR, spatial working memory, and hippocampal dendritic spine density in LH mice, and in NLH mice treated with PLX5622 consistent with a pro-cognitive role for IL-10. CONCLUSIONS Altogether these data demonstrate the critical role of IL-10 in promoting learning and memory after learned helplessness.
Collapse
Affiliation(s)
- Ryan J Worthen
- Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, Gautier Building room 415, 1011 NW 15th Street, Miami, FL, 33136, USA
| | - Susan S Garzon Zighelboim
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Gautier Building room, 4151011 NW 15th Street, Miami, FL, 33136, USA
| | - Camila S Torres Jaramillo
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Gautier Building room, 4151011 NW 15th Street, Miami, FL, 33136, USA
| | - Eleonore Beurel
- Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, Gautier Building room 415, 1011 NW 15th Street, Miami, FL, 33136, USA.
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Gautier Building room, 4151011 NW 15th Street, Miami, FL, 33136, USA.
| |
Collapse
|
75
|
Chen X, Chen H, He Y, Fu S, Liu H, Wang Q, Shen J. Proteomics-Guided Study on Buyang Huanwu Decoction for Its Neuroprotective and Neurogenic Mechanisms for Transient Ischemic Stroke: Involvements of EGFR/PI3K/Akt/Bad/14-3-3 and Jak2/Stat3/Cyclin D1 Signaling Cascades. Mol Neurobiol 2020; 57:4305-4321. [PMID: 32700252 DOI: 10.1007/s12035-020-02016-y] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 07/08/2020] [Indexed: 01/22/2023]
Abstract
Buyang Huanwu Decoction (BHD), a classic traditional Chinese medicine (TCM) formula, has been used for recovering neurological dysfunctions and treating post-stroke disability in China for 200 years. In the present study, we investigated the effects of BHD on inhibiting neuronal apoptosis, promoting proliferation and differentiation of neural stem cells (NSCs) and neurite formation and enhancing learning and memory functional recovery in an experimental rat ischemic stroke model. BHD significantly reduced infarct volume and decreased cell apoptosis in the ischemic brain. BHD enhanced neuronal cell viability in vitro. BHD dose-dependently promoted the proliferation of NSCs in ischemic rat brains in vivo. Moreover, BHD promoted neuronal and astrocyte differentiation in primary cultured NSCs in vitro. Water maze test revealed that BHD promoted the recovery of learning function but not memory functions in the transient ischemic rats. We then investigated the changes of the cellular signaling molecules by using two-dimension (2D) gel electrophoresis and focused on the PI3K/Akt/Bad and Jak2/Stat3/cyclin D1signaling pathway to uncover its underlying mechanisms for its neuroprotective and neurogenetic effects. BHD significantly upregulated the expression of p-PI3K, p-Akt, and p-Bad as well as the expression of p-Jak, p-Stat3, and cyclin D1 in vitro and in vivo. In addition, BHD upregulated Hes1 and downregulated cav-1 in vitro and in vivo. Taken together, these results suggest that BHD has neuroprotective effects and neurogenesis-promoting effects via activating PI3K/Akt/Bad and Jak2/Stat3/Cyclin D1 signaling pathways. Graphical Abstract Buyang Huanwu Decoction (BHD) activates the PI3K-AKT-BAD pathway in the ischemic brain for neuroprotection. BHD also activates JAK2/STAT3/Cyclin D1 signaling cascades for promoting neurogenesis in the hippocampus of post-ischemic brains. Moreover, BHD inhibits the expression of caveolin-1 and increases the expression of HES1 for promoting neuronal differentiation. The neuroprotective and neurogenesis-promoting effects in the hippocampus of post-ischemic brains promote learning ability.
Collapse
Affiliation(s)
- Xi Chen
- Department of Core Facility, The People's Hospital of Bao-an Shenzhen, Shenzhen, China.,The 8th people's Hospital of Shenzhen, The Affiliated Bao-an Hospital of Southern Medical University, Shenzhen, 518000, China.,School of Chinese Medicine, The University of Hong Kong, 10 Sassoon Road, Pokfulam, Hong Kong, Hong Kong SAR, China
| | - Hansen Chen
- School of Chinese Medicine, The University of Hong Kong, 10 Sassoon Road, Pokfulam, Hong Kong, Hong Kong SAR, China
| | - Yachong He
- School of Chinese Medicine, The University of Hong Kong, 10 Sassoon Road, Pokfulam, Hong Kong, Hong Kong SAR, China
| | - Shuping Fu
- School of Chinese Medicine, The University of Hong Kong, 10 Sassoon Road, Pokfulam, Hong Kong, Hong Kong SAR, China.,Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Haosheng Liu
- Department of Core Facility, The People's Hospital of Bao-an Shenzhen, Shenzhen, China.,The 8th people's Hospital of Shenzhen, The Affiliated Bao-an Hospital of Southern Medical University, Shenzhen, 518000, China
| | - Qi Wang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiangang Shen
- Department of Core Facility, The People's Hospital of Bao-an Shenzhen, Shenzhen, China. .,The 8th people's Hospital of Shenzhen, The Affiliated Bao-an Hospital of Southern Medical University, Shenzhen, 518000, China. .,School of Chinese Medicine, The University of Hong Kong, 10 Sassoon Road, Pokfulam, Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
76
|
Wertz MH, Pineda SS, Lee H, Kulicke R, Kellis M, Heiman M. Interleukin-6 deficiency exacerbates Huntington's disease model phenotypes. Mol Neurodegener 2020; 15:29. [PMID: 32448329 PMCID: PMC7247164 DOI: 10.1186/s13024-020-00379-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 05/13/2020] [Indexed: 01/04/2023] Open
Abstract
Huntington’s disease (HD) is an incurable neurodegenerative disorder caused by CAG trinucleotide expansions in the huntingtin gene. Markers of both systemic and CNS immune activation and inflammation have been widely noted in HD and mouse models of HD. In particular, elevation of the pro-inflammatory cytokine interleukin-6 (IL-6) is the earliest reported marker of immune activation in HD, and this elevation has been suggested to contribute to HD pathogenesis. To test the hypothesis that IL-6 deficiency would be protective against the effects of mutant huntingtin, we generated R6/2 HD model mice that lacked IL-6. Contrary to our prediction, IL-6 deficiency exacerbated HD-model associated behavioral phenotypes. Single nuclear RNA Sequencing (snRNA-seq) analysis of striatal cell types revealed that IL-6 deficiency led to the dysregulation of various genes associated with synaptic function, as well as the BDNF receptor Ntrk2. These data suggest that IL-6 deficiency exacerbates the effects of mutant huntingtin through dysregulation of genes of known relevance to HD pathobiology in striatal neurons, and further suggest that modulation of IL-6 to a level that promotes proper regulation of genes associated with synaptic function may hold promise as an HD therapeutic target.
Collapse
Affiliation(s)
- Mary H Wertz
- Picower Institute for Learning and Memory, Cambridge, MA, 02139, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - S Sebastian Pineda
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.,MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, MA, 02139, USA.,Department of Electrical Engineering and Computer Science, MIT, Cambridge, MA, 02139, USA
| | - Hyeseung Lee
- Picower Institute for Learning and Memory, Cambridge, MA, 02139, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Ruth Kulicke
- Picower Institute for Learning and Memory, Cambridge, MA, 02139, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Manolis Kellis
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.,MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, MA, 02139, USA.,Department of Electrical Engineering and Computer Science, MIT, Cambridge, MA, 02139, USA
| | - Myriam Heiman
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, 02139, USA. .,Picower Institute for Learning and Memory, Cambridge, MA, 02139, USA. .,Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.
| |
Collapse
|
77
|
Leptin stimulates synaptogenesis in hippocampal neurons via KLF4 and SOCS3 inhibition of STAT3 signaling. Mol Cell Neurosci 2020; 106:103500. [PMID: 32438059 DOI: 10.1016/j.mcn.2020.103500] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 03/25/2020] [Accepted: 05/05/2020] [Indexed: 01/26/2023] Open
Abstract
Normal development of neuronal connections in the hippocampus requires neurotrophic signals, including the cytokine leptin. During neonatal development, leptin induces formation and maturation of dendritic spines, the main sites of glutamatergic synapses in the hippocampal neurons. However, the molecular mechanisms for leptin-induced synaptogenesis are not entirely understood. In this study, we reveal two novel targets of leptin in developing hippocampal neurons and address their role in synaptogenesis. First target is Kruppel-Like Factor 4 (KLF4), which we identified using a genome-wide target analysis strategy. We show that leptin upregulates KLF4 in hippocampal neurons and that leptin signaling is important for KLF4 expression in vivo. Furthermore, KLF4 is required for leptin-induced synaptogenesis, as shKLF4 blocks and upregulation of KLF4 phenocopies it. We go on to show that KLF4 requires its signal transducer and activator of transcription 3 (STAT3) binding site and thus potentially blocks STAT3 activity to induce synaptogenesis. Second, we show that leptin increases the expression of suppressor of cytokine signaling 3 (SOCS3), another well-known inhibitor of STAT3, in developing hippocampal neurons. SOCS3 is also required for leptin-induced synaptogenesis and sufficient to stimulate it alone. Finally, we show that constitutively active STAT3 blocks the effects of leptin on spine formation, while the targeted knockdown of STAT3 is sufficient to induce it. Overall, our data demonstrate that leptin increases the expression of both KLF4 and SOCS3, inhibiting the activity of STAT3 in the hippocampal neurons and resulting in the enhancement of glutamatergic synaptogenesis during neonatal development.
Collapse
|
78
|
Lee SY, Song MY, Kim D, Park C, Park DK, Kim DG, Yoo JS, Kim YH. A Proteotranscriptomic-Based Computational Drug-Repositioning Method for Alzheimer's Disease. Front Pharmacol 2020; 10:1653. [PMID: 32063857 PMCID: PMC7000455 DOI: 10.3389/fphar.2019.01653] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 12/17/2019] [Indexed: 12/24/2022] Open
Abstract
Numerous clinical trials of drug candidates for Alzheimer’s disease (AD) have failed, and computational drug repositioning approaches using omics data have been proposed as effective alternative approaches to the discovery of drug candidates. However, little multi-omics data is available for AD, due to limited availability of brain tissues. Even if omics data exist, systematic drug repurposing study for AD has suffered from lack of big data, insufficient clinical information, and difficulty in data integration on account of sample heterogeneity derived from poor diagnosis or shortage of qualified post-mortem tissue. In this study, we developed a proteotranscriptomic-based computational drug repositioning method named Drug Repositioning Perturbation Score/Class (DRPS/C) based on inverse associations between disease- and drug-induced gene and protein perturbation patterns, incorporating pharmacogenomic knowledge. We constructed a Drug-induced Gene Perturbation Signature Database (DGPSD) comprised of 61,019 gene signatures perturbed by 1,520 drugs from the Connectivity Map (CMap) and the L1000 CMap. Drugs were classified into three DRPCs (High, Intermediate, and Low) according to DRPSs that were calculated using drug- and disease-induced gene perturbation signatures from DGPSD and The Cancer Genome Atlas (TCGA), respectively. The DRPS/C method was evaluated using the area under the ROC curve, with a prescribed drug list from TCGA as the gold standard. Glioblastoma had the highest AUC. To predict anti-AD drugs, DRPS were calculated using DGPSD and AD-induced gene/protein perturbation signatures generated from RNA-seq, microarray and proteomic datasets in the Synapse database, and the drugs were classified into DRPCs. We predicted 31 potential anti-AD drug candidates commonly belonged to high DRPCs of transcriptomic and proteomic signatures. Of these, four drugs classified into the nervous system group of Anatomical Therapeutic Chemical (ATC) system are voltage-gated sodium channel blockers (bupivacaine, topiramate) and monamine oxidase inhibitors (selegiline, iproniazid), and their mechanism of action was inferred from a potential anti-AD drug perspective. Our approach suggests a shortcut to discover new efficacy of drugs for AD.
Collapse
Affiliation(s)
- Soo Youn Lee
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju, South Korea
| | - Min-Young Song
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju, South Korea
| | - Dain Kim
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju, South Korea
| | - Chaewon Park
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju, South Korea
| | - Da Kyeong Park
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju, South Korea
| | - Dong Geun Kim
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju, South Korea.,Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon, South Korea
| | - Jong Shin Yoo
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju, South Korea.,Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon, South Korea
| | - Young Hye Kim
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju, South Korea
| |
Collapse
|
79
|
Girotti M, Silva JD, George CM, Morilak DA. Ciliary neurotrophic factor signaling in the rat orbitofrontal cortex ameliorates stress-induced deficits in reversal learning. Neuropharmacology 2019; 160:107791. [PMID: 31553898 DOI: 10.1016/j.neuropharm.2019.107791] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 09/19/2019] [Accepted: 09/21/2019] [Indexed: 11/17/2022]
Abstract
Deficits in cognitive flexibility, i.e. the ability to modify behavior in response to changes in the environment, are present in several psychiatric disorders and are often refractory to treatment. However, improving treatment response has been hindered by a lack of understanding of the neurobiology of cognitive flexibility. Using a rat model of chronic stress (chronic intermittent cold stress, CIC) that produces selective deficits in reversal learning, a form of cognitive flexibility dependent on orbitofrontal cortex (OFC) function, we have previously shown that JAK2 signaling is required for optimal reversal learning. In this study we explore the molecular basis of those effects. We show that, within the OFC, CIC stress reduces the levels of phosphorylated JAK2 and of ciliary neurotrophic factor (CNTF), a promoter of neuronal survival and an activator of JAK2 signaling, and that neutralizing endogenous CNTF with an intra-OFC microinjection of a specific antibody is sufficient to produce reversal-learning deficits similar to stress. Intra-OFC delivery of recombinant CNTF to CIC-stressed rats, at a dose that induces JAK2 and Akt but not STAT3 or ERK, ameliorates reversal-learning deficits, and Akt blockade prevents the positive effects of CNTF. Further analysis revealed that CNTF may exert its beneficial effects by inhibiting GSK3β, a substrate of Akt and a regulator of protein degradation. We also revealed a novel mechanism of CNTF action through modulation of p38/Mnk1/eIF4E signaling. This cascade controls translation of select mRNAs, including those encoding several plasticity-related proteins. Thus, we suggest that CNTF-driven JAK2 signaling corrects stress-induced reversal learning deficits by modulating the steady-state levels of plasticity-related proteins in the OFC.
Collapse
Affiliation(s)
- Milena Girotti
- Department of Pharmacology, Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA.
| | - Jeri D Silva
- Department of Pharmacology, Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Christina M George
- Department of Pharmacology, Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - David A Morilak
- Department of Pharmacology, Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA; South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
| |
Collapse
|
80
|
Kang M, Lee YS. The impact of RASopathy-associated mutations on CNS development in mice and humans. Mol Brain 2019; 12:96. [PMID: 31752929 PMCID: PMC6873535 DOI: 10.1186/s13041-019-0517-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 10/28/2019] [Indexed: 01/04/2023] Open
Abstract
The RAS signaling pathway is involved in the regulation of developmental processes, including cell growth, proliferation, and differentiation, in the central nervous system (CNS). Germline mutations in the RAS signaling pathway genes are associated with a group of neurodevelopmental disorders, collectively called RASopathy, which includes neurofibromatosis type 1, Noonan syndrome, cardio-facio-cutaneous syndrome, and Costello syndrome. Most mutations associated with RASopathies increase the activity of the RAS-ERK signaling pathway, and therefore, most individuals with RASopathies share common phenotypes, such as a short stature, heart defects, facial abnormalities, and cognitive impairments, which are often accompanied by abnormal CNS development. Recent studies using mouse models of RASopathies demonstrated that particular mutations associated with each disorder disrupt CNS development in a mutation-specific manner. Here, we reviewed the recent literatures that investigated the developmental role of RASopathy-associated mutations using mutant mice, which provided insights into the specific contribution of RAS-ERK signaling molecules to CNS development and the subsequent impact on cognitive function in adult mice.
Collapse
Affiliation(s)
- Minkyung Kang
- Department of Physiology, Seoul National University College of Medicine, 103 Daehak-ro, Jongro-gu, Seoul, 03080, South Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Yong-Seok Lee
- Department of Physiology, Seoul National University College of Medicine, 103 Daehak-ro, Jongro-gu, Seoul, 03080, South Korea. .,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea. .,Neuroscience Research Institute, Seoul National University College of Medicine, 103 Daehak-ro, Jongro-gu, Seoul, 03080, South Korea.
| |
Collapse
|
81
|
Privitera L, Hogg EL, Gaestel M, Wall MJ, Corrêa SAL. The MK2 cascade regulates mGluR-dependent synaptic plasticity and reversal learning. Neuropharmacology 2019; 155:121-130. [PMID: 31129151 DOI: 10.1016/j.neuropharm.2019.05.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 05/17/2019] [Accepted: 05/18/2019] [Indexed: 12/29/2022]
Abstract
The ability to either erase or update the memories of a previously learned spatial task is an essential process that is required to modify behaviour in a changing environment. Current evidence suggests that the neural representation of such cognitive flexibility involves the balancing of synaptic potentiation (acquisition of memories) with synaptic depression (modulation and updating previously acquired memories). Here we demonstrate that the p38 MAPK/MAPK-activated protein kinase 2 (MK2) cascade is required to maintain the precise tuning of long-term potentiation and long-term depression at CA1 synapses of the hippocampus which is correlated with efficient reversal learning. Using the MK2 knockout (KO) mouse, we show that mGluR-LTD, but not NMDAR-LTD, is markedly impaired in mice aged between 4 and 5 weeks (juvenile) to 7 months (mature adult). Although the amplitude of LTP was the same as in wildtype mice, priming of LTP by the activation of group I metabotropic receptors was impaired in MK2 KO mice. Consistent with unaltered LTP amplitude and compromised mGluR-LTD, MK2 KO mice had intact spatial learning when performing the Barnes maze task, but showed specific deficits in selecting the most efficient combination of search strategies to perform the task reversal. Findings from this study suggest that the mGluR-p38-MK2 cascade is important for cognitive flexibility by regulating LTD amplitude and the priming of LTP.
Collapse
Affiliation(s)
- Lucia Privitera
- (a)Bradford School of Pharmacy and Medical Sciences, University of Bradford, Bradford, BD7 1DP, United Kingdom
| | - Ellen L Hogg
- (a)Bradford School of Pharmacy and Medical Sciences, University of Bradford, Bradford, BD7 1DP, United Kingdom
| | - Matthias Gaestel
- Institute of Cell Biochemistry, Hannover Medical University, 30625, Hannover, Germany
| | - Mark J Wall
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, United Kingdom
| | - Sonia A L Corrêa
- (a)Bradford School of Pharmacy and Medical Sciences, University of Bradford, Bradford, BD7 1DP, United Kingdom.
| |
Collapse
|
82
|
Shariq AS, Brietzke E, Rosenblat JD, Pan Z, Rong C, Ragguett RM, Park C, McIntyre RS. Therapeutic potential of JAK/STAT pathway modulation in mood disorders. Rev Neurosci 2019; 30:1-7. [PMID: 29902157 DOI: 10.1515/revneuro-2018-0027] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 04/11/2018] [Indexed: 12/20/2022]
Abstract
Convergent evidence demonstrates that immune dysfunction (e.g. chronic low-grade inflammatory activation) plays an important role in the development and progression of mood disorders. The Janus kinase/signal transducers and activators of transcription (JAK/STAT) signaling pathway is a pleiotropic cellular cascade that transduces numerous signals, including signals from the release of cytokines and growth factors. The JAK/STAT signaling pathway is involved in mediating several functions of the central nervous system, including neurogenesis, synaptic plasticity, gliogenesis, and microglial activation, all of which have been implicated in the pathophysiology of mood disorders. In addition, the antidepressant actions of current treatments have been shown to be mediated by JAK/STAT-dependent mechanisms. To date, two JAK inhibitors (JAKinibs) have been approved by the U.S. Food and Drug Administration and are primarily indicated for the treatment of inflammatory conditions such as rheumatoid arthritis. Indirect evidence from studies in populations with inflammatory conditions indicates that JAKinibs significantly improve measures of mood and quality of life. There is also direct evidence from studies in populations with depressive disorders, suggesting that JAK/STAT pathways may be involved in the pathophysiology of depression and that the inhibition of specific JAK/STAT pathways (i.e. via JAKinibs) may be a promising novel treatment for depressive disorders.
Collapse
Affiliation(s)
- Aisha S Shariq
- Mood Disorders Psychopharmacology Unit (MDPU), Toronto Western Hospital, University Health Network (UHN), Toronto, Ontario M5T 2S8, Canada
| | - Elisa Brietzke
- Mood Disorders Psychopharmacology Unit (MDPU), Toronto Western Hospital, University Health Network (UHN), Toronto, Ontario M5T 2S8, Canada.,Research Group in Molecular and Behavioral Neuroscience of Bipolar Disorder, Department of Psychiatry, Federal University of São Paulo (UNIFESP), São Paulo 14021-001, Brazil
| | - Joshua D Rosenblat
- Mood Disorders Psychopharmacology Unit (MDPU), Toronto Western Hospital, University Health Network (UHN), Toronto, Ontario M5T 2S8, Canada.,Department of Psychiatry, University of Toronto, Toronto, Ontario M5S 1A1, Canada
| | - Zihang Pan
- Mood Disorders Psychopharmacology Unit (MDPU), Toronto Western Hospital, University Health Network (UHN), Toronto, Ontario M5T 2S8, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario M5S 1A1, Canada
| | - Carola Rong
- Mood Disorders Psychopharmacology Unit (MDPU), Toronto Western Hospital, University Health Network (UHN), Toronto, Ontario M5T 2S8, Canada
| | - Renee-Marie Ragguett
- Mood Disorders Psychopharmacology Unit (MDPU), Toronto Western Hospital, University Health Network (UHN), Toronto, Ontario M5T 2S8, Canada
| | - Caroline Park
- Mood Disorders Psychopharmacology Unit (MDPU), Toronto Western Hospital, University Health Network (UHN), Toronto, Ontario M5T 2S8, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario M5S 1A1, Canada
| | - Roger S McIntyre
- Mood Disorders Psychopharmacology Unit (MDPU), Toronto Western Hospital, University Health Network (UHN), Toronto, Ontario M5T 2S8, Canada.,Department of Psychiatry, University of Toronto, Toronto, Ontario M5S 1A1, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario M5S 1A1, Canada.,Brain and Cognition Discovery Foundation (BCDF), Toronto, Ontario M5T 2S8, Canada
| |
Collapse
|
83
|
Hixson KM, Cogswell M, Brooks-Kayal AR, Russek SJ. Evidence for a non-canonical JAK/STAT signaling pathway in the synthesis of the brain's major ion channels and neurotransmitter receptors. BMC Genomics 2019; 20:677. [PMID: 31455240 PMCID: PMC6712773 DOI: 10.1186/s12864-019-6033-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 08/15/2019] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Brain-derived neurotrophic factor (BDNF) is a major signaling molecule that the brain uses to control a vast network of intracellular cascades fundamental to properties of learning and memory, and cognition. While much is known about BDNF signaling in the healthy nervous system where it controls the mitogen activated protein kinase (MAPK) and cyclic-AMP pathways, less is known about its role in multiple brain disorders where it contributes to the dysregulated neuroplasticity seen in epilepsy and traumatic brain injury (TBI). We previously found that neurons respond to prolonged BDNF exposure (both in vivo (in models of epilepsy and TBI) and in vitro (in BDNF treated primary neuronal cultures)) by activating the Janus Kinase/Signal Transducer and Activator of Transcription (JAK/STAT) signaling pathway. This pathway is best known for its association with inflammatory cytokines in non-neuronal cells. RESULTS Here, using deep RNA-sequencing of neurons exposed to BDNF in the presence and absence of well characterized JAK/STAT inhibitors, and without non-neuronal cells, we determine the BDNF transcriptome that is specifically regulated by agents that inhibit JAK/STAT signaling. Surprisingly, the BDNF-induced JAK/STAT transcriptome contains ion channels and neurotransmitter receptors coming from all the major classes expressed in the brain, along with key modulators of synaptic plasticity, neurogenesis, and axonal remodeling. Analysis of this dataset has revealed a unique non-canonical mechanism of JAK/STATs in neurons as differential gene expression mediated by STAT3 is not solely dependent upon phosphorylation at residue 705 and may involve a BDNF-induced interaction of STAT3 with Heterochromatin Protein 1 alpha (HP1α). CONCLUSIONS These findings suggest that the neuronal BDNF-induced JAK/STAT pathway involves more than STAT3 phosphorylation at 705, providing the first evidence for a non-canonical mechanism that may involve HP1α. Our analysis reveals that JAK/STAT signaling regulates many of the genes associated with epilepsy syndromes where BDNF levels are markedly elevated. Uncovering the mechanism of this novel form of BDNF signaling in the brain may provide a new direction for epilepsy therapeutics and open a window into the complex mechanisms of STAT3 transcriptional regulation in neurological disease.
Collapse
Affiliation(s)
- Kathryn M. Hixson
- Laboratory of Translational Epilepsy, Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine (BUSM), Boston, USA
- Graduate Program for Neuroscience (GPN), Boston University (BU), Boston, USA
| | - Meaghan Cogswell
- Laboratory of Translational Epilepsy, Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine (BUSM), Boston, USA
| | - Amy R. Brooks-Kayal
- Department of Pediatric Neurology, University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Shelley J. Russek
- Laboratory of Translational Epilepsy, Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine (BUSM), Boston, USA
- Graduate Program for Neuroscience (GPN), Boston University (BU), Boston, USA
- Department of Biology, Boston University (BU), Boston, USA
| |
Collapse
|
84
|
Chen Q, Lv J, Yang W, Xu B, Wang Z, Yu Z, Wu J, Yang Y, Han Y. Targeted inhibition of STAT3 as a potential treatment strategy for atherosclerosis. Theranostics 2019; 9:6424-6442. [PMID: 31588227 PMCID: PMC6771242 DOI: 10.7150/thno.35528] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 07/10/2019] [Indexed: 02/06/2023] Open
Abstract
Atherosclerosis is the main pathological basis of ischemic cardiovascular and cerebrovascular diseases and has attracted more attention in recent years. Multiple studies have demonstrated that the signal transducer and activator of transcription 3 (STAT3) plays essential roles in the process of atherosclerosis. Moreover, aberrant STAT3 activation has been shown to contribute to the occurrence and development of atherosclerosis. Therefore, the study of STAT3 inhibitors has gradually become a focal research topic. In this review, we describe the crucial roles of STAT3 in endothelial cell dysfunction, macrophage polarization, inflammation, and immunity during atherosclerosis. STAT3 in mitochondria is mentioned as well. Then, we present a summary and classification of STAT3 inhibitors, which could offer potential treatment strategies for atherosclerosis. Furthermore, we enumerate some of the problems that have interfered with the development of mature therapies utilizing STAT3 inhibitors to treat atherosclerosis. Finally, we propose ideas that may help to solve these problems to some extent. Collectively, this review may be useful for developing future STAT3 inhibitor therapies for atherosclerosis.
Collapse
|
85
|
Kobylarek D, Iwanowski P, Lewandowska Z, Limphaibool N, Szafranek S, Labrzycka A, Kozubski W. Advances in the Potential Biomarkers of Epilepsy. Front Neurol 2019; 10:685. [PMID: 31312171 PMCID: PMC6614180 DOI: 10.3389/fneur.2019.00685] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 06/12/2019] [Indexed: 12/11/2022] Open
Abstract
Epilepsy is a group of chronic neurological disorders characterized by recurrent, spontaneous, and unpredictable seizures. It is one of the most common neurological disorders, affecting tens of millions of people worldwide. Comprehensive studies on epilepsy in recent decades have revealed the complexity of epileptogenesis, in which immunological processes, epigenetic modifications, and structural changes in neuronal tissues have been identified as playing a crucial role. This review discusses the recent advances in the biomarkers of epilepsy. We evaluate the possible molecular background underlying the clinical changes observed in recent studies, focusing on therapeutic investigations, and the evidence of their safety and efficacy in the human population. This article reviews the pathophysiology of epilepsy, including recent reports on the effects of oxidative stress and hypoxia, and focuses on specific biomarkers and their clinical implications, along with further perspectives in epilepsy research.
Collapse
Affiliation(s)
- Dominik Kobylarek
- Department of Neurology, Poznan University of Medical Sciences, Poznan, Poland
| | | | | | | | | | | | | |
Collapse
|
86
|
Irisin Attenuates Neuroinflammation and Prevents the Memory and Cognitive Deterioration in Streptozotocin-Induced Diabetic Mice. Mediators Inflamm 2019; 2019:1567179. [PMID: 31281225 PMCID: PMC6590589 DOI: 10.1155/2019/1567179] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 03/30/2019] [Accepted: 05/13/2019] [Indexed: 12/11/2022] Open
Abstract
Diabetes mellitus (DM) patients experience memory and cognitive deficits. The mechanisms underlying this dysfunction in the brain of DM patients are not fully understood, and therefore, no optimized therapeutic strategy has been established so far. The aim of the present study was to assess whether irisin was able to improve memory and cognitive performance in a streptozotocin-induced diabetic mouse model. A diabetic mouse model was established and behavioral tests were performed. We also set up primary cultures for mechanism studies. Western blots and EMSA were used for molecular studies. Significant impairment of cognition and memory was observed in these DM mice, which could be effectively prevented by irisin cotreatment. We also found upregulated levels of GFAP protein, reduced synaptic protein expression, and increased levels of interleukin-1β (IL-1β) and interleukin-6 (IL-6) in the brains; however, irisin significantly attenuated these cellular responses. Meanwhile, our results demonstrated that irisin inhibited the activation of P38, STAT3, and NFκB proteins of DM mice. Furthermore, our results suggested that irisin might regulate the function of P38, STAT3, and NFκB in hippocampal tissues of DM mice. Collectively, irisin inhibited neuroinflammation in STZ-induced DM mice by inhibiting cytokine release and improving their cognitive function. Our findings revealed the mechanism of irisin's anti-inflammatory effect in the CNS.
Collapse
|
87
|
Yang L, Luo M, He J, Zuo H, Weng S, He J, Xu X. A JAK-STAT pathway target gene encoding a single WAP domain (SWD)-containing protein from Litopenaeus vannamei. FISH & SHELLFISH IMMUNOLOGY 2019; 89:555-563. [PMID: 30999041 DOI: 10.1016/j.fsi.2019.04.046] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 04/11/2019] [Accepted: 04/12/2019] [Indexed: 06/09/2023]
Abstract
In shrimp, the JAK-STAT pathway is essentially implicated in both antiviral and antibacterial responses. However, few regulatory target genes of the JAK-STAT pathway in shrimp have been reported so far. In this study, a novel single WAP domain-containing peptide (LvSWD4) was identified from Pacific white shrimp Litopenaeus vannamei. The promoter of LvSWD4 was predicted to harbor multiple STAT-binding DNA motifs. Over-expression of the JAK-STAT pathway components STAT, JAK and Domeless in vitro significantly enhanced the transcriptional activity of the LvSWD4 promoter, and in vivo silencing of STAT and the the JAK-STAT pathway upstream regulator IRF down-regulated the expression of LvSWD4, suggesting that LvSWD4 could be a target gene of the JAK-STAT pathway. The expression of LvSWD4 was significantly increased after infection with Gram-negative and positive bacteria, fungi and virus, and silencing of LvSWD4 increased the susceptibility of shrimp to V. parahaemolyticus and WSSV infections. In vitro experiments also demonstrated that the recombinant LvSWD4 protein had significant inhibitory activities against Gram negative bacteria V. parahaemolyticus and E. coli and Gram positive bacteria S. aureus and B. subtilis. Furthermore, silencing of LvSWD4 in vivo significantly affected expression of various immune functional genes and attenuated the phagocytic activity of hemocytes. These suggested that as a target gene of STAT, LvSWD4 was essentially implicated in shrimp immunity, which could constitute part of the mechanism underlying the immune function of the shrimp JAK-STAT pathway.
Collapse
Affiliation(s)
- Linwei Yang
- MOE Key Laboratory of Aquatic Product Safety, State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China; Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, PR China
| | - Mengting Luo
- MOE Key Laboratory of Aquatic Product Safety, State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China; Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, PR China
| | - Jianhui He
- MOE Key Laboratory of Aquatic Product Safety, State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China; Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, PR China
| | - Hongliang Zuo
- MOE Key Laboratory of Aquatic Product Safety, State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China; Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, PR China
| | - Shaoping Weng
- MOE Key Laboratory of Aquatic Product Safety, State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China; Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, PR China
| | - Jianguo He
- MOE Key Laboratory of Aquatic Product Safety, State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China; Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, PR China; South China Sea Resource Exploitation and Protection Collaborative Innovation Center (SCS-REPIC), Guangzhou, PR China
| | - Xiaopeng Xu
- MOE Key Laboratory of Aquatic Product Safety, State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China; Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, PR China; South China Sea Resource Exploitation and Protection Collaborative Innovation Center (SCS-REPIC), Guangzhou, PR China.
| |
Collapse
|
88
|
Mango D, Nisticò R. Acid-Sensing Ion Channel 1a Is Involved in N-Methyl D-Aspartate Receptor-Dependent Long-Term Depression in the Hippocampus. Front Pharmacol 2019; 10:555. [PMID: 31178731 PMCID: PMC6537656 DOI: 10.3389/fphar.2019.00555] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 05/02/2019] [Indexed: 11/29/2022] Open
Abstract
Acid-sensing ion channels (ASICs), members of the degenerin/epithelial Na+ channel superfamily, are largely expressed in the mammalian nervous system. ASIC1a is highly permeable to Ca2+ and are involved in many physiological processes, including synaptic plasticity, learning, and memory. To clarify the role of ASIC1a in synaptic transmission and plasticity, we investigated N-methyl D-aspartate (NMDA) receptor-dependent long-term depression (LTD) in the CA1 region of the hippocampus. We found that: (1) ASIC1a mediates a component of ASIC1a excitatory postsynaptic currents (EPSCs); (2) ASIC1a plays a role in electrical LTD induced by LFS protocol both in P13-18 and P30-40 animals; (3) ASIC1a is involved in chemical LTD induced by brief bath application of NMDA both in P13-18 and P30-40 animals; and finally (4) a functional interaction between ASIC1a and NMDA receptors occurs during LTD. These findings suggest a new role for ASIC1a in specific forms of synaptic plasticity in the mouse hippocampus.
Collapse
Affiliation(s)
- D Mango
- Laboratory of Neuropharmacology, European Brain Research Institute, Rita Levi-Montalcini Foundation, Rome, Italy
| | - R Nisticò
- Laboratory of Neuropharmacology, European Brain Research Institute, Rita Levi-Montalcini Foundation, Rome, Italy.,Department of Biology, School of Pharmacy, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
89
|
Nevado-Holgado AJ, Ribe E, Thei L, Furlong L, Mayer MA, Quan J, Richardson JC, Cavanagh J, Consortium N, Lovestone S. Genetic and Real-World Clinical Data, Combined with Empirical Validation, Nominate Jak-Stat Signaling as a Target for Alzheimer's Disease Therapeutic Development. Cells 2019; 8:cells8050425. [PMID: 31072055 PMCID: PMC6562942 DOI: 10.3390/cells8050425] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 04/29/2019] [Accepted: 05/01/2019] [Indexed: 11/16/2022] Open
Abstract
As genome-wide association studies (GWAS) have grown in size, the number of genetic variants that have been associated per disease has correspondingly increased. Despite this increase in the number of single-nucleotide polymorphisms (SNPs) identified per disease, their biological interpretation has in many cases remained elusive. To address this, we have combined GWAS results with orthogonal sources of evidence, namely the current knowledge of molecular pathways; real-world clinical data from six million patients; RNA expression across tissues from Alzheimer’s disease (AD) patients, and purpose-built rodent models for experimental validation. In more detail, first we show that when examined at a pathway level, analysis of all GWAS studies groups AD in a cluster with disorders of immunity and inflammation. Using clinical data, we show that the degree of comorbidity of these diseases with AD correlates with the strength of their genetic association with molecular participants in the Janus kinases/signal transducer and activator of transcription (JAK-STAT) pathway. Using four independent RNA expression datasets we then find evidence for the altered regulation of JAK-STAT pathway genes in AD. Finally, we use both in vitro and in vivo rodent models to demonstrate that Aβ induces gene expression of the key drivers of this pathway, providing experimental evidence to validate these data-driven observations. These results therefore nominate JAK-STAT anomalies as a prominent aetiopathological event in AD and hence a potential target for therapeutic development, and moreover demonstrate a de novo multi-modal approach to derive information from rapidly increasing genomic datasets.
Collapse
Affiliation(s)
- Alejo J Nevado-Holgado
- Department of Psychiatry, University of Oxford, Oxford OX3 7JX, UK.
- Oxford Health NHS Foundation Trust, Oxford OX3 7JX, UK.
| | - Elena Ribe
- Department of Psychiatry, University of Oxford, Oxford OX3 7JX, UK.
| | - Laura Thei
- Department of Psychiatry, University of Oxford, Oxford OX3 7JX, UK.
| | - Laura Furlong
- GRIB, Hospital del Mar Medical Research Institute, 08003 Barcelona, Spain.
| | - Miguel-Angel Mayer
- GRIB, Hospital del Mar Medical Research Institute, 08003 Barcelona, Spain.
| | - Jie Quan
- Computational Sciences, Worldwide Research and Development, Pfizer Inc. 1 Portland St, Cambridge MA 02139, USA.
| | - Jill C Richardson
- Neurosciences Therapeutic Area Unit, Glaxo Smith Kline R&D Ltd., Stevenage SG1 2NY, UK.
| | - Jonathan Cavanagh
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, G12 8TA UK.
| | | | - Simon Lovestone
- Department of Psychiatry, University of Oxford, Oxford OX3 7JX, UK.
| |
Collapse
|
90
|
Moskalev A, Proshkina E, Zhavoronkov A, Shaposhnikov M. Effects of unpaired 1 gene overexpression on the lifespan of Drosophila melanogaster. BMC SYSTEMS BIOLOGY 2019; 13:16. [PMID: 30836998 PMCID: PMC6399815 DOI: 10.1186/s12918-019-0687-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Background The JAK/STAT signaling pathway is involved in many aging-related cellular functions. However, effects of overexpression of genes controlling JAK/STAT signal transduction on longevity of model organisms have not been studied. Here we evaluate the effect of overexpression of the unpaired 1 (upd1) gene, which encodes an activating ligand for JAK/STAT pathway, on the lifespan of Drosophila melanogaster. Results Overexpression of upd1 in the intestine caused a pronounced shortening of the median lifespan by 54.1–18.9%, and the age of 90% mortality by 40.9–19.1% in males and females, respectively. In fat body and in nervous system of male flies, an induction of upd1 overexpression increased the age of 90% mortality and median lifespan, respectively. An increase in upd1 expression enhanced mRNA levels of the JAK/STAT target genes domeless and Socs36E. Conclusions Conditional overexpression of upd1 in different tissues of Drosophila imago induces pro-aging or pro-longevity effects in tissue-dependent manner. The effects of upd1 overexpression on lifespan are accompanied by the transcription activation of genes for the components of JAK/STAT pathway.
Collapse
Affiliation(s)
- Alexey Moskalev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia. .,Institute of Biology, Komi Scientific Center, Ural Division, Russian Academy of Sciences, Syktyvkar, 167982, Russia. .,Moscow Institute of Physics and Technology, Dolgoprudny, 141700, Russia.
| | - Ekaterina Proshkina
- Institute of Biology, Komi Scientific Center, Ural Division, Russian Academy of Sciences, Syktyvkar, 167982, Russia
| | | | - Mikhail Shaposhnikov
- Institute of Biology, Komi Scientific Center, Ural Division, Russian Academy of Sciences, Syktyvkar, 167982, Russia
| |
Collapse
|
91
|
Xie G, Song C, Lin X, Yang M, Fan X, Liu W, Tao J, Chen L, Huang J. Electroacupuncture Regulates Hippocampal Synaptic Plasticity via Inhibiting Janus-Activated Kinase 2/Signal Transducer and Activator of Transcription 3 Signaling in Cerebral Ischemic Rats. J Stroke Cerebrovasc Dis 2019; 28:792-799. [DOI: 10.1016/j.jstrokecerebrovasdis.2018.11.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 10/18/2018] [Accepted: 11/22/2018] [Indexed: 01/16/2023] Open
|
92
|
McGregor G, Harvey J. Leptin Regulation of Synaptic Function at Hippocampal TA-CA1 and SC-CA1 Synapses: Implications for Health and Disease. Neurochem Res 2019; 44:650-660. [PMID: 28819795 PMCID: PMC6420429 DOI: 10.1007/s11064-017-2362-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 07/05/2017] [Accepted: 07/21/2017] [Indexed: 12/16/2022]
Abstract
Growing evidence indicates that the endocrine hormone leptin regulates hippocampal synaptic function in addition to its established role as a hypothalamic satiety signal. Indeed, numerous studies show that leptin facilitates the cellular events that underlie hippocampal learning and memory including activity-dependent synaptic plasticity and glutamate receptor trafficking, indicating that leptin may be a potential cognitive enhancer. Although there has been extensive investigation into the modulatory role of leptin at hippocampal Schaffer collateral (SC)-CA1 synapses, recent evidence indicates that leptin also potently regulates excitatory synaptic transmission at the anatomically distinct temporoammonic (TA) input to hippocampal CA1 neurons. The cellular mechanisms underlying activity-dependent synaptic plasticity at TA-CA1 synapses differ from those at SC-CA1 synapses and the TA input is implicated in spatial and episodic memory formation. Furthermore, the TA input is an early target for neurodegeneration in Alzheimer's disease (AD) and aberrant leptin function is linked to AD. Here, we review the evidence that leptin regulates hippocampal synaptic function at both SC- and TA-CA1 synapses and discuss the consequences for neurodegenerative disorders like AD.
Collapse
Affiliation(s)
- Gemma McGregor
- Division of Neuroscience, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK
| | - Jenni Harvey
- Division of Neuroscience, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK.
| |
Collapse
|
93
|
Lee HC, Md Yusof HH, Leong MPY, Zainal Abidin S, Seth EA, Hewitt CA, Vidyadaran S, Nordin N, Scott HS, Cheah PS, Ling KH. Gene and protein expression profiles of JAK-STAT signalling pathway in the developing brain of the Ts1Cje down syndrome mouse model. Int J Neurosci 2019; 129:871-881. [DOI: 10.1080/00207454.2019.1580280] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Han-Chung Lee
- Genetics and Regenerative Medicine Research Centre (GRMRC), Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
| | - Hadri Hadi Md Yusof
- Genetics and Regenerative Medicine Research Centre (GRMRC), Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
| | - Melody Pui-Yee Leong
- Genetics and Regenerative Medicine Research Centre (GRMRC), Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
| | - Shahidee Zainal Abidin
- Genetics and Regenerative Medicine Research Centre (GRMRC), Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
| | - Eryse Amira Seth
- Genetics and Regenerative Medicine Research Centre (GRMRC), Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Chelsee A. Hewitt
- Department of Pathology, The Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - Sharmili Vidyadaran
- Genetics and Regenerative Medicine Research Centre (GRMRC), Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
- Department of Pathology, Immunology Unit, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Norshariza Nordin
- Genetics and Regenerative Medicine Research Centre (GRMRC), Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
| | - Hamish S. Scott
- Department of Genetics and Molecular Pathology, Centre for Cancer Biology, An alliance between SA Pathology and the University of South Australia, SA Pathology, Adelaide, Australia
- School of Pharmacy and Medical Science, University of South Australia, Adelaide, Australia
- School of Medicine, School of Biological Sciences, University of Adelaide, Adelaide, South Australia
- Centre for Cancer Biology, SA Pathology, Australian Cancer Research Foundation Genomics Facility, Adelaide, Australia
| | - Pike-See Cheah
- Genetics and Regenerative Medicine Research Centre (GRMRC), Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - King-Hwa Ling
- Genetics and Regenerative Medicine Research Centre (GRMRC), Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
| |
Collapse
|
94
|
Menezes GD, Faria-Melibeu AC, Serfaty CA, Campello-Costa P. In vivo effect of acute exposure to interleukin-6 on the developing visual system. Neurosci Lett 2019; 698:7-12. [PMID: 30611891 DOI: 10.1016/j.neulet.2019.01.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 12/10/2018] [Accepted: 01/02/2019] [Indexed: 01/12/2023]
Abstract
Interleukin-6 (IL-6) is involved in different processes of the central nervous system. Our aims were to investigate the effect of IL-6 on retinotectal topography and on different signaling pathways. Rats were submitted to an intravitreous injection of either IL-6 (50 ng/ml) or PBS (vehicle) at postnatal day 10 (PND10). At PND11 or PND14, different groups were processed for western blot, histochemistry or immunofluorescence analysis. IL-6 treatment leads to an increase in pSTAT-3 levels in the retina and a disruption in the retinotectal topographic map, suggesting that a transient increase in interleukin-6 levels may impact neural circuitry development.
Collapse
Affiliation(s)
- Grasielle Duarte Menezes
- Programa de Neurociências, Departamento de Neurobiologia, Universidade Federal Fluminense, Niterói, RJ, Brazil.
| | - Adriana C Faria-Melibeu
- Programa de Neurociências, Departamento de Neurobiologia, Universidade Federal Fluminense, Niterói, RJ, Brazil.
| | - Claudio Alberto Serfaty
- Programa de Neurociências, Departamento de Neurobiologia, Universidade Federal Fluminense, Niterói, RJ, Brazil.
| | - Paula Campello-Costa
- Programa de Neurociências, Departamento de Neurobiologia, Universidade Federal Fluminense, Niterói, RJ, Brazil.
| |
Collapse
|
95
|
Venuto S, Castellana S, Monti M, Appolloni I, Fusilli C, Fusco C, Pucci P, Malatesta P, Mazza T, Merla G, Micale L. TRIM8-driven transcriptomic profile of neural stem cells identified glioma-related nodal genes and pathways. Biochim Biophys Acta Gen Subj 2018; 1863:491-501. [PMID: 30528352 DOI: 10.1016/j.bbagen.2018.12.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 11/26/2018] [Accepted: 12/03/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND We recently reported TRIM8, encoding an E3 ubiquitin ligase, as a gene aberrantly expressed in glioblastoma whose expression suppresses cell growth and induces a significant reduction of clonogenic potential in glioblastoma cell lines. METHODS we provided novel insights on TRIM8 functions by profiling the transcriptome of TRIM8-expressing primary mouse embryonal neural stem cells by RNA-sequencing and bioinformatic analysis. Functional analysis including luciferase assay, western blot, PCR arrays, Real time quantitative PCR were performed to validate the transcriptomic data. RESULTS Our study identified enriched pathways related to the neurotransmission and to the central nervous system (CNS) functions, including axonal guidance, GABA receptor, Ephrin B, synaptic long-term potentiation/depression, and glutamate receptor signalling pathways. Finally, we provided additional evidence about the existence of a functional interactive crosstalk between TRIM8 and STAT3. CONCLUSIONS Our results substantiate the role of TRIM8 in the brain functions through the dysregulation of genes involved in different CNS-related pathways, including JAK-STAT. GENERAL SIGNIFICANCE This study provides novel insights on the physiological TRIM8 function by profiling for the first time the primary Neural Stem Cell over-expressing TRIM8 by using RNA-Sequencing methodology.
Collapse
Affiliation(s)
- Santina Venuto
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale Padre Pio, 71013, San Giovanni Rotondo, Foggia, Italy; Experimental and Regenerative Medicine, University of Foggia, Via A. Gramsci, 89/91, 71122, Foggia, Italy.
| | - Stefano Castellana
- Bioinformatics Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale Padre Pio, 71013, San Giovanni Rotondo, Foggia, Italy.
| | - Maria Monti
- CEINGE Advanced Biotechnology, Department of Chemical Sciences, Federico II University, Via Gaetano Salvatore, 486, 80145, Napoli, Italy.
| | - Irene Appolloni
- U.O. Medicina Rigenerativa Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genova, Italy
| | - Caterina Fusilli
- Bioinformatics Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale Padre Pio, 71013, San Giovanni Rotondo, Foggia, Italy.
| | - Carmela Fusco
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale Padre Pio, 71013, San Giovanni Rotondo, Foggia, Italy.
| | - Piero Pucci
- CEINGE Advanced Biotechnology, Department of Chemical Sciences, Federico II University, Via Gaetano Salvatore, 486, 80145, Napoli, Italy.
| | - Paolo Malatesta
- U.O. Medicina Rigenerativa Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genova, Italy; Department of Experimental Medicine (DiMES), University of Genova, Via Leon Battista Alberti, 2, 16132 Genova, Italy.
| | - Tommaso Mazza
- Bioinformatics Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale Padre Pio, 71013, San Giovanni Rotondo, Foggia, Italy.
| | - Giuseppe Merla
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale Padre Pio, 71013, San Giovanni Rotondo, Foggia, Italy.
| | - Lucia Micale
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale Padre Pio, 71013, San Giovanni Rotondo, Foggia, Italy.
| |
Collapse
|
96
|
Bliim N, Leshchyns'ka I, Keable R, Chen BJ, Curry-Hyde A, Gray L, Sytnyk V, Janitz M. Early transcriptome changes in response to chemical long-term potentiation induced via activation of synaptic NMDA receptors in mouse hippocampal neurons. Genomics 2018; 111:1676-1686. [PMID: 30465913 DOI: 10.1016/j.ygeno.2018.11.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 11/07/2018] [Accepted: 11/16/2018] [Indexed: 01/23/2023]
Abstract
Long term potentiation (LTP) is a form of synaptic plasticity. In the present study LTP was induced via activation of synaptic NMDA receptors in primary hippocampal neuron cultures from neonate mice and RNA was isolated for RNA sequencing at 20 min following LTP induction. RNA sequencing and differential expression testing was performed to determine the identity and abundance of protein-coding and non-coding RNAs in control and LTP induced neuron cultures. We show that expression levels of a small group of transcripts encoding proteins involved in negative regulation of gene expression (Adcyap1, Id3), protein translation (Rpl22L1), extracellular structure organization (Bgn), intracellular signalling (Ppm1H, Ntsr2, Cldn10) and protein citrullination (PAD2) are downregulated in the stimulated neurons. Our results suggest that the early stages of LTP are accompanied by the remodelling of the biosynthetic machinery, interactions with the extracellular matrix and intracellular signalling pathways at the transcriptional level.
Collapse
Affiliation(s)
- Nicola Bliim
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Iryna Leshchyns'ka
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Ryan Keable
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Bei Jun Chen
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Ashton Curry-Hyde
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Lachlan Gray
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Vladimir Sytnyk
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia.
| | - Michael Janitz
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia; Paul-Flechsig-Institute for Brain Research, University of Leipzig, Leipzig, Germany.
| |
Collapse
|
97
|
Del Olmo N, Ruiz-Gayo M. Influence of High-Fat Diets Consumed During the Juvenile Period on Hippocampal Morphology and Function. Front Cell Neurosci 2018; 12:439. [PMID: 30515083 PMCID: PMC6255817 DOI: 10.3389/fncel.2018.00439] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 11/05/2018] [Indexed: 12/18/2022] Open
Abstract
The negative impact of obesity on neurocognitive functioning is an issue of increasing clinical interest. Over the last decade, a number of studies have analyzed the influence of high-fat diets (HFDs) on cognitive performance, particularly in adolescent individuals. Different approaches, including behavioral, neurochemical, electrophysiological and morphological studies, have been developed to address the effect of HFDs on neural processes interfering with learning and memory skills in rodents. Many of the studies have focused on learning and memory related to the hippocampus and the mechanisms underlying these processes. The goal of the current review article is to highlight the relationship between hippocampal learning/memory deficits and nutritional/endocrine inputs derived from HFDs consumption, with a special emphasis on research showing the effect of HFDs intake during the juvenile period. We have also reviewed recent research regarding the effect of HFDs on hippocampal neurotransmission. An overview of research suggesting the involvement of fatty acid (FA) receptor-mediated signaling pathways in memory deficits triggered by HFDs is also provided. Finally, the role of leptin and HFD-evoked hyperleptinemia is discussed.
Collapse
Affiliation(s)
- Nuria Del Olmo
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, Madrid, Spain
| | - Mariano Ruiz-Gayo
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, Madrid, Spain
| |
Collapse
|
98
|
Varenicline reduces DNA damage, tau mislocalization and post surgical cognitive impairment in aged mice. Neuropharmacology 2018; 143:217-227. [PMID: 30273594 DOI: 10.1016/j.neuropharm.2018.09.044] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 09/02/2018] [Accepted: 09/26/2018] [Indexed: 12/20/2022]
Abstract
Postoperative cognitive dysfunction (POCD) occurs more frequently in elderly patients undergoing major surgery. Age associated cholinergic imbalance may exacerbate postoperative systemic and neuroinflammation, but the effect nicotinic acetylcholine receptor (nAchR) stimulation on the development of POCD remains unclear. Aged male C57BL/6N mice (18 months old) underwent a midline laparotomy or were exposed to sevoflurane anesthesia alone (4-5%), with or without concomitant varenicline, a partial nAchR, at 1 mg/kg/day. Laparotomy increased pro-inflammatory cytokines in the liver and hippocampus (IL-1β and MCP-1) and induced a decline in cognitive performance, indicated by lower discrimination index in the Novel Object Recognition test, greater error number and longer escape latency in the Y-maze test. Glia activation, aberrant tau phosphorylation (AT8) and accumulation of phosphorylated H2AX in the hippocampus were detectable up to postoperative day 14, with neuronal apoptosis seen in the hippocampus. Perioperative varenicline attenuated the cognitive decline and associated tau protein mislocalization, DNA damage and neuronal apoptosis. The modulation of JAK2/STAT3 signaling may play a critical role in this process. Neuroinflammation, tau phosphorylation and DNA damage contribute to the development of cognitive dysfunction following laparotomy. Cholinergic stimulation by varenicline attenuated these changes through preventing the mislocalization of phosphorylated tau and DNA damage.
Collapse
|
99
|
du Plessis N, Kotze LA, Leukes V, Walzl G. Translational Potential of Therapeutics Targeting Regulatory Myeloid Cells in Tuberculosis. Front Cell Infect Microbiol 2018; 8:332. [PMID: 30298121 PMCID: PMC6160538 DOI: 10.3389/fcimb.2018.00332] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 08/28/2018] [Indexed: 12/11/2022] Open
Abstract
Despite recent advances in tuberculosis (TB) drug development and availability, successful antibiotic treatment is challenged by the parallel development of antimicrobial resistance. As a result, new approaches toward improving TB treatment have been proposed in an attempt to reduce the high TB morbidity and mortality rates. Host-directed therapies (HDTs), designed to modulate host immune components, provide an alternative approach for improving treatment outcome in both non-communicable and infectious diseases. Many candidate immunotherapeutics, designed to target regulatory myeloid immune components in cancer, have so far proven to be of value as repurposed HDT in TB. Several of these studies do however lack detailed description of the mechanism or host pathway affected by TB HDT treatment. In this review, we present an argument for greater appreciation of the role of regulatory myeloid cells, such as myeloid-derived suppressor cells (MDSC), as potential targets for the development of candidate TB HDT compounds. We discuss the role of MDSC in the context of Mycobacterium tuberculosis infection and disease, focussing primarily on their specific cellular functions and highlight the impact of HDTs on MDSC frequency and function.
Collapse
Affiliation(s)
- Nelita du Plessis
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | - Leigh A Kotze
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | - Vinzeigh Leukes
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | - Gerhard Walzl
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
100
|
McGregor G, Clements L, Farah A, Irving AJ, Harvey J. Age-dependent regulation of excitatory synaptic transmission at hippocampal temporoammonic-CA1 synapses by leptin. Neurobiol Aging 2018; 69:76-93. [PMID: 29860205 PMCID: PMC6075472 DOI: 10.1016/j.neurobiolaging.2018.05.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 04/17/2018] [Accepted: 05/04/2018] [Indexed: 12/15/2022]
Abstract
The hippocampus is a key target for the hormone leptin and leptin regulation of excitatory synaptic transmission at Schaffer-collateral-CA1 synapses during aging are well documented. However, little is known about the age-dependent actions of leptin at the temporoammonic (TA) input to CA1 neurons. Here we show that leptin induces a novel form of N-methyl-D-aspartate receptor-dependent long-term depression (LTD) at adult (12-24 weeks old) TA-CA1 synapses. Leptin-induced LTD requires activation of canonical Janus tyrosine kinase 2- signal transducer and activator of transcription signaling and removal of GluA1-containing α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors from synapses. Moreover, leptin-induced LTD is occluded by activity-dependent LTD at TA-CA1 synapses. By contrast, leptin has no effect on excitatory synaptic transmission at aged (12-14 months old) TA-CA1 synapses, and low-frequency stimulation also fails to induce LTD at this age. These findings demonstrate clear age-related alterations in the leptin sensitivity of TA-CA1 synapses and provide valuable information on how the leptin system alters with age. As leptin has been linked to Alzheimer's disease, these findings have important implications for understanding of age-related disorders such as Alzheimer's disease.
Collapse
Affiliation(s)
- Gemma McGregor
- Division of Neuroscience, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Leigh Clements
- Division of Neuroscience, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Adham Farah
- Division of Neuroscience, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Andrew J Irving
- School of Biomolecular and Biomedical Science, The Conway Institute, University College Dublin, Dublin, Ireland
| | - Jenni Harvey
- Division of Neuroscience, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK.
| |
Collapse
|