1
|
Guo Q, Li ZF, Hu DY, Li PJ, Wu KN, Fan HH, Deng J, Wu HM, Zhang X, Zhu JH. The selenocysteine-containing protein SELENOT maintains dopamine signaling in the midbrain to protect mice from hyperactivity disorder. EMBO J 2025:10.1038/s44318-025-00430-3. [PMID: 40195499 DOI: 10.1038/s44318-025-00430-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/10/2025] [Accepted: 03/14/2025] [Indexed: 04/09/2025] Open
Abstract
Dopaminergic neuron dysfunction has been implicated in multiple neurological and psychiatric disorders. SELENOT is a selenocysteine-containing protein of the ER membrane with antioxidant and neuroprotective activities, but its pathophysiological role in dopaminergic neurons remains unclear. In this study we show that male mice with SELENOT-deficient dopaminergic neurons exhibit attention deficit/hyperactivity disorder (ADHD)-like symptoms, including hyperlocomotion, recognition memory deficits, repetitive movements, and impulsivity. Dopamine metabolism, extrasynaptic dopamine levels, spontaneous excitatory postsynaptic currents in the striatum, and electroencephalography theta power are all enhanced in these animals, while dopaminergic neurons in the substantia nigra are slightly reduced but with normal firing and cellular stress levels. Our results also indicate that the expression of dopamine transporter (DAT) is significantly reduced in the absence of SELENOT. Both the development of ADHD-like phenotypes and DAT downregulation are also observed when SELENOT is absent from the whole brain, but not when its conditional knockout is restricted to astrocytes. Mechanistically, we show that SELENOT downregulates DAT expression via interaction with SERCA2 of the ER -but not with IP3R or RYR- to regulate the ER-cytosol Ca2+ flux and, subsequently, the activity of transcription factor NURR1 and the expression levels of DAT. Treatment with amphetamine or methylphenidate, which are commonly used to treat ADHD, reverses the hyperactivity observed in mice with SELENOT-deficient dopaminergic neurons. Our study demonstrates that SELENOT in mouse dopaminergic neurons maintains proper dopamine signaling in the midbrain against the development of ADHD-like behaviors.
Collapse
Affiliation(s)
- Qing Guo
- Institute of Nutrition and Diseases and Center for Research, School of Public Health, Wenzhou Medical University, Wenzhou, China
- Department of Neurology and Institute of Geriatric Neurology, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Zhao-Feng Li
- Institute of Nutrition and Diseases and Center for Research, School of Public Health, Wenzhou Medical University, Wenzhou, China
| | - Dong-Yan Hu
- Institute of Nutrition and Diseases and Center for Research, School of Public Health, Wenzhou Medical University, Wenzhou, China
| | - Pei-Jun Li
- Department of Neurology and Institute of Geriatric Neurology, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Kai-Nian Wu
- Institute of Nutrition and Diseases and Center for Research, School of Public Health, Wenzhou Medical University, Wenzhou, China
| | - Hui-Hui Fan
- Institute of Nutrition and Diseases and Center for Research, School of Public Health, Wenzhou Medical University, Wenzhou, China
| | - Jie Deng
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Hong-Mei Wu
- Institute of Nutrition and Diseases and Center for Research, School of Public Health, Wenzhou Medical University, Wenzhou, China
| | - Xiong Zhang
- Department of Neurology and Institute of Geriatric Neurology, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China.
| | - Jian-Hong Zhu
- Institute of Nutrition and Diseases and Center for Research, School of Public Health, Wenzhou Medical University, Wenzhou, China.
- Department of Neurology and Institute of Geriatric Neurology, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
2
|
Li K, Feng Z, Xiong Z, Pan J, Zhou M, Li W, Ou Y, Wu G, Che M, Gong H, Peng J, Wang X, Qi S, Peng J. Growth hormone promotes the reconstruction of injured axons in the hypothalamo-neurohypophyseal system. Neural Regen Res 2024; 19:2249-2258. [PMID: 38488559 PMCID: PMC11034602 DOI: 10.4103/1673-5374.389358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 07/11/2023] [Accepted: 09/14/2023] [Indexed: 04/24/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202410000-00026/figure1/v/2024-02-06T055622Z/r/image-tiff Previous studies have shown that growth hormone can regulate hypothalamic energy metabolism, stress, and hormone release. Therefore, growth hormone has great potential for treating hypothalamic injury. In this study, we established a specific hypothalamic axon injury model by inducing hypothalamic pituitary stalk electric lesions in male mice. We then treated mice by intraperitoneal administration of growth hormone. Our results showed that growth hormone increased the expression of insulin-like growth factor 1 and its receptors, and promoted the survival of hypothalamic neurons, axonal regeneration, and vascular reconstruction from the median eminence through the posterior pituitary. Altogether, this alleviated hypothalamic injury-caused central diabetes insipidus and anxiety. These results suggest that growth hormone can promote axonal reconstruction after hypothalamic injury by regulating the growth hormone-insulin-like growth factor 1 axis.
Collapse
Affiliation(s)
- Kai Li
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Zhanpeng Feng
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Zhiwei Xiong
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Jun Pan
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Mingfeng Zhou
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Weizhao Li
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Yichao Ou
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Guangsen Wu
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Mengjie Che
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Haodong Gong
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Junjie Peng
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Xingqin Wang
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Songtao Qi
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Junxiang Peng
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| |
Collapse
|
3
|
Ma ZL, Wang ZL, Zhang FY, Liu HX, Mao LH, Yuan L. Biomarkers of Parkinson's Disease: From Basic Research to Clinical Practice. Aging Dis 2024; 15:1813-1830. [PMID: 37815899 PMCID: PMC11272192 DOI: 10.14336/ad.2023.1005] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/05/2023] [Indexed: 10/12/2023] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease characterized pathologically by dopaminergic neuron loss and the formation of Lewy bodies, which are enriched with aggregated α-synuclein (α-syn). PD currently has no cure, but therapeutic strategies are available to alleviate symptoms. Early diagnosis can greatly improve therapeutic interventions, but the clinical diagnosis of PD remains challenging and depends mainly on clinical features and imaging tests. Efficient and specific biomarkers are crucial for the diagnosis, monitoring, and evaluation of PD. Here, we reviewed the biomarkers of PD in different tissues and biofluids, along with the current clinical biochemical detection methods. We found that the sensitivity and specificity of single biomarkers are limited, and selecting appropriate indicators for combined detection can improve the diagnostic accuracy of PD.
Collapse
Affiliation(s)
| | | | - Fei-yue Zhang
- Laboratory of Research in Parkinson’s Disease and Related Disorders, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute, China Medical University, Shenyang, China
| | - Hong-xun Liu
- Laboratory of Research in Parkinson’s Disease and Related Disorders, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute, China Medical University, Shenyang, China
| | - Li-hong Mao
- Laboratory of Research in Parkinson’s Disease and Related Disorders, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute, China Medical University, Shenyang, China
| | - Lin Yuan
- Laboratory of Research in Parkinson’s Disease and Related Disorders, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute, China Medical University, Shenyang, China
| |
Collapse
|
4
|
Zhao X, Zou H, Wang M, Wang J, Wang T, Wang L, Chen X. Conformal Neuromorphic Bioelectronics for Sense Digitalization. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2403444. [PMID: 38934554 DOI: 10.1002/adma.202403444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/03/2024] [Indexed: 06/28/2024]
Abstract
Sense digitalization, the process of transforming sensory experiences into digital data, is an emerging research frontier that links the physical world with human perception and interaction. Inspired by the adaptability, fault tolerance, robustness, and energy efficiency of biological senses, this field drives the development of numerous innovative digitalization techniques. Neuromorphic bioelectronics, characterized by biomimetic adaptability, stand out for their seamless bidirectional interactions with biological entities through stimulus-response and feedback loops, incorporating bio-neuromorphic intelligence for information exchange. This review illustrates recent progress in sensory digitalization, encompassing not only the digital representation of physical sensations such as touch, light, and temperature, correlating to tactile, visual, and thermal perceptions, but also the detection of biochemical stimuli such as gases, ions, and neurotransmitters, mirroring olfactory, gustatory, and neural processes. It thoroughly examines the material design, device manufacturing, and system integration, offering detailed insights. However, the field faces significant challenges, including the development of new device/system paradigms, forging genuine connections with biological systems, ensuring compatibility with the semiconductor industry and overcoming the absence of standardization. Future ambition includes realization of biocompatible neural prosthetics, exoskeletons, soft humanoid robots, and cybernetic devices that integrate smoothly with both biological tissues and artificial components.
Collapse
Affiliation(s)
- Xiao Zhao
- State Key Laboratory of Organic Electronics and Information Displays, Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing, 210023, China
| | - Haochen Zou
- State Key Laboratory of Organic Electronics and Information Displays, Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing, 210023, China
| | - Ming Wang
- Frontier Institute of Chip and System, State Key Laboratory of Integrated Chips and Systems, Fudan University, Shanghai, 200433, China
| | - Jianwu Wang
- Institute for Digital Molecular Analytics and Science (IDMxS), Nanyang Technological University, 59 Nanyang Drive, Singapore, 636921, Singapore
- Innovative Centre for Flexible Devices (iFLEX) Max Planck-NTU Joint Lab for Artificial Senses, School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Ting Wang
- State Key Laboratory of Organic Electronics and Information Displays, Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing, 210023, China
| | - Lianhui Wang
- State Key Laboratory of Organic Electronics and Information Displays, Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing, 210023, China
| | - Xiaodong Chen
- Institute for Digital Molecular Analytics and Science (IDMxS), Nanyang Technological University, 59 Nanyang Drive, Singapore, 636921, Singapore
- Innovative Centre for Flexible Devices (iFLEX) Max Planck-NTU Joint Lab for Artificial Senses, School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| |
Collapse
|
5
|
Wang M, Pugh SM, Daboul J, Miller D, Xu Y, Hill JW. IGF-1 Acts through Kiss1-expressing Cells to Influence Metabolism and Reproduction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.02.601722. [PMID: 39005405 PMCID: PMC11244982 DOI: 10.1101/2024.07.02.601722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Objective Kisspeptin, encoded by the Kiss1 gene, ties puberty and fertility to energy status; however, the metabolic factors that control Kiss1-expressing cells need to be clarified. Methods To evaluate the impact of IGF-1 on the metabolic and reproductive functions of kisspeptin producing cells, we created mice with IGF-1 receptor deletion driven by the Kiss1 promoter (IGF1RKiss1 mice). Previous studies have shown IGF-1 and insulin can bind to each other's receptor, permitting IGF-1 signaling in the absence of IGF1R. Therefore, we also generated mice with simultaneous deletion of the IGF1R and insulin receptor (IR) in Kiss1-expressing cells (IGF1R/IRKiss1 mice). Results Loss of IGF1R in Kiss1 cells caused stunted body length. In addition, female IGF1RKiss1 mice displayed lower body weight and food intake plus higher energy expenditure and physical activity. This phenotype was linked to higher proopiomelanocortin (POMC) expression and heightened brown adipose tissue (BAT) thermogenesis. Male IGF1RKiss1 mice had mild changes in metabolic functions. Moreover, IGF1RKiss1 mice of both sexes experienced delayed puberty. Notably, male IGF1RKiss1 mice had impaired adulthood fertility accompanied by lower gonadotropin and testosterone levels. Thus, IGF1R in Kiss1-expressing cells impacts metabolism and reproduction in a sex-specific manner. IGF1R/IRKiss1 mice had higher fat mass and glucose intolerance, suggesting IGF1R and IR in Kiss1-expressing cells together regulate body composition and glucose homeostasis. Conclusions Overall, our study shows that IGF1R and IR in Kiss1 have cooperative roles in body length, metabolism, and reproduction.
Collapse
Affiliation(s)
- Mengjie Wang
- Center for Diabetes and Endocrine Research, University of Toledo College of Medicine, Toledo, Ohio, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Seamus M. Pugh
- Center for Diabetes and Endocrine Research, University of Toledo College of Medicine, Toledo, Ohio, USA
| | - Judy Daboul
- Center for Diabetes and Endocrine Research, University of Toledo College of Medicine, Toledo, Ohio, USA
| | - David Miller
- Center for Diabetes and Endocrine Research, University of Toledo College of Medicine, Toledo, Ohio, USA
| | - Yong Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Jennifer W. Hill
- Center for Diabetes and Endocrine Research, University of Toledo College of Medicine, Toledo, Ohio, USA
- Department of Obstetrics and Gynecology, University of Toledo College of Medicine, Toledo, Ohio, USA
| |
Collapse
|
6
|
Tan MMX, Lawton MA, Pollard MI, Brown E, Real R, Carrasco AM, Bekadar S, Jabbari E, Reynolds RH, Iwaki H, Blauwendraat C, Kanavou S, Hubbard L, Malek N, Grosset KA, Bajaj N, Barker RA, Burn DJ, Bresner C, Foltynie T, Wood NW, Williams-Gray CH, Andreassen OA, Toft M, Elbaz A, Artaud F, Brice A, Corvol JC, Aasly J, Farrer MJ, Nalls MA, Singleton AB, Williams NM, Ben-Shlomo Y, Hardy J, Hu MTM, Grosset DG, Shoai M, Pihlstrøm L, Morris HR. Genome-wide determinants of mortality and motor progression in Parkinson's disease. NPJ Parkinsons Dis 2024; 10:113. [PMID: 38849413 PMCID: PMC11161485 DOI: 10.1038/s41531-024-00729-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 05/24/2024] [Indexed: 06/09/2024] Open
Abstract
There are 90 independent genome-wide significant genetic risk variants for Parkinson's disease (PD) but currently only five nominated loci for PD progression. The biology of PD progression is likely to be of central importance in defining mechanisms that can be used to develop new treatments. We studied 6766 PD patients, over 15,340 visits with a mean follow-up of between 4.2 and 15.7 years and carried out genome-wide survival studies for time to a motor progression endpoint, defined by reaching Hoehn and Yahr stage 3 or greater, and death (mortality). There was a robust effect of the APOE ε4 allele on mortality in PD. We also identified a locus within the TBXAS1 gene encoding thromboxane A synthase 1 associated with mortality in PD. We also report 4 independent loci associated with motor progression in or near MORN1, ASNS, PDE5A, and XPO1. Only the non-Gaucher disease causing GBA1 PD risk variant E326K, of the known PD risk variants, was associated with mortality in PD. Further work is needed to understand the links between these genomic variants and the underlying disease biology. However, these may represent new candidates for disease modification in PD.
Collapse
Affiliation(s)
- Manuela M X Tan
- Department of Neurology, Oslo University Hospital, Oslo, Norway.
- Department of Clinical and Movement Neurosciences, Queen Square Institute of Neurology, University College London, London, UK.
- UCL Movement Disorders Centre, University College London, London, UK.
| | - Michael A Lawton
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Miriam I Pollard
- Department of Clinical and Movement Neurosciences, Queen Square Institute of Neurology, University College London, London, UK
| | - Emmeline Brown
- Department of Clinical and Movement Neurosciences, Queen Square Institute of Neurology, University College London, London, UK
| | - Raquel Real
- Department of Clinical and Movement Neurosciences, Queen Square Institute of Neurology, University College London, London, UK
- UCL Movement Disorders Centre, University College London, London, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Alejandro Martinez Carrasco
- Department of Clinical and Movement Neurosciences, Queen Square Institute of Neurology, University College London, London, UK
- UCL Movement Disorders Centre, University College London, London, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Samir Bekadar
- Sorbonne University, Paris Brain Institute - ICM, Inserm, CNRS, Assistance Publique Hôpitaux de Paris, Departement of Neurology, Hôpital Pitié-Salpêtrière, Paris, France
| | - Edwin Jabbari
- Department of Clinical and Movement Neurosciences, Queen Square Institute of Neurology, University College London, London, UK
- UCL Movement Disorders Centre, University College London, London, UK
| | - Regina H Reynolds
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Hirotaka Iwaki
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica, Washington DC, USA
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, USA
| | - Cornelis Blauwendraat
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, USA
| | - Sofia Kanavou
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Leon Hubbard
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | - Naveed Malek
- Department of Neurology, Institute of Neurological Sciences, Queen Elizabeth University Hospital, Glasgow, UK
| | - Katherine A Grosset
- Department of Neurology, Institute of Neurological Sciences, Queen Elizabeth University Hospital, Glasgow, UK
| | - Nin Bajaj
- Clinical Neurosciences, University of Nottingham, Nottingham, UK
| | - Roger A Barker
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - David J Burn
- Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Catherine Bresner
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | - Thomas Foltynie
- Department of Clinical and Movement Neurosciences, Queen Square Institute of Neurology, University College London, London, UK
- UCL Movement Disorders Centre, University College London, London, UK
| | - Nicholas W Wood
- Department of Clinical and Movement Neurosciences, Queen Square Institute of Neurology, University College London, London, UK
- UCL Movement Disorders Centre, University College London, London, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Caroline H Williams-Gray
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Ole A Andreassen
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Mathias Toft
- Department of Neurology, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Alexis Elbaz
- Paris-Saclay University, UVSQ, Inserm, Gustave Roussy, "Exposome and Heredity" team, CESP, 94807, Villejuif, France
| | - Fanny Artaud
- Paris-Saclay University, UVSQ, Inserm, Gustave Roussy, "Exposome and Heredity" team, CESP, 94807, Villejuif, France
| | - Alexis Brice
- Sorbonne University, Paris Brain Institute - ICM, Inserm, CNRS, Assistance Publique Hôpitaux de Paris, Departement of Neurology, Hôpital Pitié-Salpêtrière, Paris, France
| | - Jean-Christophe Corvol
- Sorbonne University, Paris Brain Institute - ICM, Inserm, CNRS, Assistance Publique Hôpitaux de Paris, Departement of Neurology, Hôpital Pitié-Salpêtrière, Paris, France
| | - Jan Aasly
- Department of Neurology, St. Olavs Hospital, Trondheim, Norway
- Department of Neuromedicine and Movement Science (INB), Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Matthew J Farrer
- Department of Neurology, University of Florida, Gainesville, FL, USA
| | - Michael A Nalls
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica, Washington DC, USA
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, USA
| | - Andrew B Singleton
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, USA
| | - Nigel M Williams
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | - Yoav Ben-Shlomo
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - John Hardy
- UCL Movement Disorders Centre, University College London, London, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Department of Neurodegenerative Diseases, Queen Square Institute of Neurology, University College London, London, UK
- Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute, University College London, London, UK
- National Institute for Health Research (NIHR) University College London Hospitals Biomedical Research Centre, London, UK
- Institute for Advanced Study, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Michele T M Hu
- Nuffield Department of Clinical Neurosciences, Division of Clinical Neurology, University of Oxford, Oxford, UK
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford, UK
- Department of Clinical Neurology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Donald G Grosset
- School of Neuroscience and Psychology, University of Glasgow, Glasgow, UK
| | - Maryam Shoai
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Department of Neurodegenerative Diseases, Queen Square Institute of Neurology, University College London, London, UK
- Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, London, UK
| | - Lasse Pihlstrøm
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Huw R Morris
- Department of Clinical and Movement Neurosciences, Queen Square Institute of Neurology, University College London, London, UK.
- UCL Movement Disorders Centre, University College London, London, UK.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA.
| |
Collapse
|
7
|
Yaghmaeian Salmani B, Lahti L, Gillberg L, Jacobsen JK, Mantas I, Svenningsson P, Perlmann T. Transcriptomic atlas of midbrain dopamine neurons uncovers differential vulnerability in a Parkinsonism lesion model. eLife 2024; 12:RP89482. [PMID: 38587883 PMCID: PMC11001297 DOI: 10.7554/elife.89482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2024] Open
Abstract
Midbrain dopamine (mDA) neurons comprise diverse cells with unique innervation targets and functions. This is illustrated by the selective sensitivity of mDA neurons of the substantia nigra compacta (SNc) in patients with Parkinson's disease, while those in the ventral tegmental area (VTA) are relatively spared. Here, we used single nuclei RNA sequencing (snRNA-seq) of approximately 70,000 mouse midbrain cells to build a high-resolution atlas of mouse mDA neuron diversity at the molecular level. The results showed that differences between mDA neuron groups could best be understood as a continuum without sharp differences between subtypes. Thus, we assigned mDA neurons to several 'territories' and 'neighborhoods' within a shifting gene expression landscape where boundaries are gradual rather than discrete. Based on the enriched gene expression patterns of these territories and neighborhoods, we were able to localize them in the adult mouse midbrain. Moreover, because the underlying mechanisms for the variable sensitivities of diverse mDA neurons to pathological insults are not well understood, we analyzed surviving neurons after partial 6-hydroxydopamine (6-OHDA) lesions to unravel gene expression patterns that correlate with mDA neuron vulnerability and resilience. Together, this atlas provides a basis for further studies on the neurophysiological role of mDA neurons in health and disease.
Collapse
Affiliation(s)
| | - Laura Lahti
- Department of Cell and Molecular Biology, Karolinska InstitutetStockholmSweden
| | - Linda Gillberg
- Department of Cell and Molecular Biology, Karolinska InstitutetStockholmSweden
| | - Jesper Kjaer Jacobsen
- Department of Cell and Molecular Biology, Karolinska InstitutetStockholmSweden
- Department of Neurology, Karolinska University HospitalStockholmSweden
| | - Ioannis Mantas
- Department of Clinical Neuroscience, Karolinska InstitutetStockholmSweden
| | - Per Svenningsson
- Department of Clinical Neuroscience, Karolinska InstitutetStockholmSweden
| | - Thomas Perlmann
- Department of Cell and Molecular Biology, Karolinska InstitutetStockholmSweden
| |
Collapse
|
8
|
Chen F, Lu K, Bai N, Hao Y, Wang H, Zhao X, Yue F. Oral administration of ellagic acid mitigates perioperative neurocognitive disorders, hippocampal oxidative stress, and neuroinflammation in aged mice by restoring IGF-1 signaling. Sci Rep 2024; 14:2509. [PMID: 38291199 PMCID: PMC10827749 DOI: 10.1038/s41598-024-53127-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 01/29/2024] [Indexed: 02/01/2024] Open
Abstract
This study investigates the potential of ellagic acid (EA), a phytochemical with antioxidant and anti-inflammatory properties, in managing perioperative neurocognitive disorders (PND). PND, which represents a spectrum of cognitive impairments often faced by elderly patients, is principally linked to surgical and anesthesia procedures, and heavily impacted by oxidative stress in the hippocampus and microglia-induced neuroinflammation. Employing an aged mice model subjected to abdominal surgery, we delve into EA's ability to counteract postoperative oxidative stress and cerebral inflammation by engaging the Insulin-like growth factor-1 (IGF-1) pathway. Our findings revealed that administering EA orally notably alleviated post-surgical cognitive decline in older mice, a fact that was manifested in improved performance during maze tests. This enhancement in the behavioral performance of the EA-treated mice corresponded with the rejuvenation of IGF-1 signaling, a decrease in oxidative stress markers in the hippocampus (like MDA and carbonylated protein), and an increase in the activity of antioxidant enzymes such as SOD and CAT. Alongside these, we observed a decrease in microglia-driven neuroinflammation in the hippocampus, thus underscoring the antioxidant and anti-inflammatory roles of EA. Interestingly, when EA was given in conjunction with an IGF1R inhibitor, these benefits were annulled, accentuating the pivotal role that the IGF-1 pathway plays in the neuroprotective potential of EA. Hence, EA could serve as a potent candidate for safeguarding against PND in older patients by curbing oxidative stress and neuroinflammation through the activation of the IGF-1 pathway.
Collapse
Affiliation(s)
- Fang Chen
- Department of Anesthesiology, Shaanxi Provincial People's Hospital, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, 710068, Shaanxi, China
| | - Kai Lu
- Department of Anesthesiology, Shaanxi Provincial People's Hospital, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, 710068, Shaanxi, China
| | - Ning Bai
- Department of Anesthesiology, Shaanxi Provincial People's Hospital, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, 710068, Shaanxi, China
| | - Yabo Hao
- Department of Anesthesiology, Shaanxi Provincial People's Hospital, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, 710068, Shaanxi, China
| | - Hui Wang
- Department of Anesthesiology, Shaanxi Provincial People's Hospital, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, 710068, Shaanxi, China
| | - Xinrong Zhao
- Department of Anesthesiology, Shaanxi Provincial People's Hospital, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, 710068, Shaanxi, China
| | - Fang Yue
- Department of Anesthesiology, Shaanxi Provincial People's Hospital, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, 710068, Shaanxi, China.
| |
Collapse
|
9
|
Cui L, Gao L, Geng H, Zhang H, Wei H. Analysis of the relationship between mild cognitive impairment and serum klotho protein and insulin-like growth factor-1 in the elderly. Technol Health Care 2024; 32:1455-1462. [PMID: 37599547 DOI: 10.3233/thc-230462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
BACKGROUND Mild cognitive impairment (MCI) is a mild memory or cognitive impairment. OBJECTIVE To explore the relationship between serum klotho (K1) protein and insulin-like growth factor-1 and mild cognitive impairment in the elderly in order to provide accurate and appropriate indicators for clinical diagnosis and treatment of MCI. METHODS This randomized stratified study adopted a multistage cluster sampling method. 161 elderly patients with mild cognitive impairment were included as the MCI group, and 161 healthy people matched with the MCI group in gender, age and education were selected as the control group. RESULTS The levels of serum K1 protein and insulin-like growth factor-1 in the MCI group were lower than those in the control group (P< 0.05). Both IGF-1 and K1 had predictive value for MCI (P< 0.05). The area under the curve (AUC) of IGF-1 for predicting MCI was 0.859 (95% CI: 0.790∼0.929), and the AUC of K1 for predicting MCI was 0.793 (95% CI: 0.694∼0.892). The value of joint prediction of the two indicators was the highest, with an AUC of 0.939 (95% CI: 0.896-0.993). CONCLUSION High serum K1 and insulin-like growth factor-1 are the protective factors of cognitive impairment in MCI patients. Both IGF-1 and serum K1 proteins have predictive value for MCI, and the combination of the two indicators has the highest predictive value.
Collapse
|
10
|
Herrero-Labrador R, Fernández-Irigoyen J, Vecino R, González-Arias C, Ausín K, Crespo I, Fernández Acosta FJ, Nieto-Estévez V, Román MJ, Perea G, Torres-Alemán I, Santamaría E, Vicario C. Brain IGF-I regulates LTP, spatial memory, and sexual dimorphic behavior. Life Sci Alliance 2023; 6:e202201691. [PMID: 37463753 PMCID: PMC10355288 DOI: 10.26508/lsa.202201691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 06/29/2023] [Accepted: 07/03/2023] [Indexed: 07/20/2023] Open
Abstract
Insulin-like growth factor-I (IGF-I) exerts multiple actions, yet the role of IGF-I from different sources is poorly understood. Here, we explored the functional and behavioral consequences of the conditional deletion of Igf-I in the nervous system (Igf-I Δ/Δ), and demonstrated that long-term potentiation was impaired in hippocampal slices. Moreover, Igf-I Δ/Δ mice showed spatial memory deficits in the Morris water maze, and the significant sex-dependent differences displayed by Igf-I Ctrl/Ctrl mice disappeared in Igf-I Δ/Δ mice in the open field and rota-rod tests. Brain Igf-I deletion disorganized the granule cell layer of the dentate gyrus (DG), and it modified the relative expressions of GAD and VGLUT1, which are preferentially localized to inhibitory and excitatory presynaptic terminals. Furthermore, Igf-I deletion altered protein modules involved in receptor trafficking, synaptic proteins, and proteins that functionally interact with estrogen and androgen metabolism. Our findings indicate that brain IGF-I is crucial for long-term potentiation, and that it is involved in the regulation of spatial memory and sexual dimorphic behaviors, possibly by maintaining the granule cell layer structure and the stability of synaptic-related protein modules.
Collapse
Affiliation(s)
- Raquel Herrero-Labrador
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- CIBERNED, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Joaquín Fernández-Irigoyen
- Proteored-ISCIII, Proteomics Platform, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Clinical Neuroproteomics Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Rebeca Vecino
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- CIBERNED, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | | | - Karina Ausín
- Proteored-ISCIII, Proteomics Platform, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Inmaculada Crespo
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- CIBERNED, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- CES Cardenal Cisneros, Madrid, Spain
| | | | - Vanesa Nieto-Estévez
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- CIBERNED, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - M José Román
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- CIBERNED, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Gertrudis Perea
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Ignacio Torres-Alemán
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- CIBERNED, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Achucarro Basque Center for Neuroscience, and Ikerbasque Foundation for Science, Bilbao, Spain
| | - Enrique Santamaría
- Proteored-ISCIII, Proteomics Platform, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Clinical Neuroproteomics Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Carlos Vicario
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- CIBERNED, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
11
|
Rune GM, Joue G, Sommer T. Effects of 24-hour oral estradiol-valerate administration on hormone levels in men and pre-menopausal women. Psychoneuroendocrinology 2023; 156:106320. [PMID: 37307791 DOI: 10.1016/j.psyneuen.2023.106320] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 06/06/2023] [Accepted: 06/07/2023] [Indexed: 06/14/2023]
Abstract
In order to translate the findings from the vast animal literature on the effect of 17β-estradiol (E2) on brain and behavior to humans, a placebo-controlled pharmacological enhancement of E2 levels for at least 24 h is necessary. However, an exogenous increase in E2 for such a prolonged period might affect the endogenous secretion of other (neuroactive) hormones. Such effects would be of relevance for the interpretation of the effects of this pharmacological regimen on cognition and its neural correlates as well as be of basic scientific interest. We therefore administered a double dose of 12 mg of estradiol-valerate (E2V) to men and of 8 mg to naturally cycling women in their low-hormone phase, and assessed the concentration of two steroids critical to hormone regulation: follicle stimulating hormone (FSH) and luteinizing hormone (LH). We also assessed any changes in concentration of the neuroactive hormones progesterone (P4), testosterone (TST), dihydrotestosterone (DHT) and immune-like growth factor 1 (IGF-1). This regimen resulted in similar E2 levels in both sexes (saliva and serum). FSH and LH levels in both sexes were down-regulated to the same degree. P4 concentration decreased in both sexes only in serum but not saliva. TST and DHT levels dropped only in men whereas sex-hormone binding globulin was not affected. Finally, the concentration of IGF-1 decreased in both sexes. Based on previous studies on the effects of these neuroactive hormones, only the degree of downregulation of TST and DHT levels in men might have an impact on brain and behavior, which should be considered when interpreting the effects of the presented E2V regimes.
Collapse
Affiliation(s)
- Gabriele M Rune
- Institute of Cell Biology and Neurobiology, Charité Anatomy, Charitéplatz 1, 10117 Berlin, Germany
| | - Gina Joue
- Institute of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, Martinistr. 52, D-20248 Hamburg, Germany
| | - Tobias Sommer
- Institute of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, Martinistr. 52, D-20248 Hamburg, Germany.
| |
Collapse
|
12
|
Tu X, Jain A, Parra Bueno P, Decker H, Liu X, Yasuda R. Local autocrine plasticity signaling in single dendritic spines by insulin-like growth factors. SCIENCE ADVANCES 2023; 9:eadg0666. [PMID: 37531435 PMCID: PMC10396292 DOI: 10.1126/sciadv.adg0666] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 06/29/2023] [Indexed: 08/04/2023]
Abstract
The insulin superfamily of peptides is essential for homeostasis as well as neuronal plasticity, learning, and memory. Here, we show that insulin-like growth factors 1 and 2 (IGF1 and IGF2) are differentially expressed in hippocampal neurons and released in an activity-dependent manner. Using a new fluorescence resonance energy transfer sensor for IGF1 receptor (IGF1R) with two-photon fluorescence lifetime imaging, we find that the release of IGF1 triggers rapid local autocrine IGF1R activation on the same spine and more than several micrometers along the stimulated dendrite, regulating the plasticity of the activated spine in CA1 pyramidal neurons. In CA3 neurons, IGF2, instead of IGF1, is responsible for IGF1R autocrine activation and synaptic plasticity. Thus, our study demonstrates the cell type-specific roles of IGF1 and IGF2 in hippocampal plasticity and a plasticity mechanism mediated by the synthesis and autocrine signaling of IGF peptides in pyramidal neurons.
Collapse
Affiliation(s)
- Xun Tu
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
- International Max Planck Research School for Brain and Behavior, Jupiter, FL, USA
- FAU/Max Planck Florida Institute Joint Graduate Program in Integrative Biology and Neuroscience, Florida Atlantic University, Boca Raton, FL, USA
| | - Anant Jain
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Paula Parra Bueno
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Helena Decker
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Xiaodan Liu
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Ryohei Yasuda
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| |
Collapse
|
13
|
Li M, Larsen PA. Single-cell sequencing of entorhinal cortex reveals widespread disruption of neuropeptide networks in Alzheimer's disease. Alzheimers Dement 2023; 19:3575-3592. [PMID: 36825405 DOI: 10.1002/alz.12979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/12/2023] [Accepted: 01/15/2023] [Indexed: 02/25/2023]
Abstract
INTRODUCTION Abnormalities of neuropeptides (NPs) that play important roles in modulating neuronal activities are commonly observed in Alzheimer's disease (AD). We hypothesize that NP network disruption is widespread in AD brains. METHODS Single-cell transcriptomic data from the entorhinal cortex (EC) were used to investigate the NP network disruption in AD. Bulk RNA-sequencing data generated from the temporal cortex by independent groups and machine learning were employed to identify key NPs involved in AD. The relationship between aging and AD-associated NP (ADNP) expression was studied using GTEx data. RESULTS The proportion of cells expressing NPs but not their receptors decreased significantly in AD. Neurons expressing higher level and greater diversity of NPs were disproportionately absent in AD. Increased age coincides with decreased ADNP expression in the hippocampus. DISCUSSION NP network disruption is widespread in AD EC. Neurons expressing more NPs may be selectively vulnerable to AD. Decreased expression of NPs participates in early AD pathogenesis. We predict that the NP network can be harnessed for treatment and/or early diagnosis of AD.
Collapse
Affiliation(s)
- Manci Li
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, USA
- Minnesota Center for Prion Research and Outreach, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA
| | - Peter A Larsen
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, USA
- Minnesota Center for Prion Research and Outreach, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA
| |
Collapse
|
14
|
Beccano-Kelly DA, Cherubini M, Mousba Y, Cramb KM, Giussani S, Caiazza MC, Rai P, Vingill S, Bengoa-Vergniory N, Ng B, Corda G, Banerjee A, Vowles J, Cowley S, Wade-Martins R. Calcium dysregulation combined with mitochondrial failure and electrophysiological maturity converge in Parkinson's iPSC-dopamine neurons. iScience 2023; 26:107044. [PMID: 37426342 PMCID: PMC10329047 DOI: 10.1016/j.isci.2023.107044] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/30/2022] [Accepted: 06/01/2023] [Indexed: 07/11/2023] Open
Abstract
Parkinson's disease (PD) is characterized by a progressive deterioration of motor and cognitive functions. Although death of dopamine neurons is the hallmark pathology of PD, this is a late-stage disease process preceded by neuronal dysfunction. Here we describe early physiological perturbations in patient-derived induced pluripotent stem cell (iPSC)-dopamine neurons carrying the GBA-N370S mutation, a strong genetic risk factor for PD. GBA-N370S iPSC-dopamine neurons show an early and persistent calcium dysregulation notably at the mitochondria, followed by reduced mitochondrial membrane potential and oxygen consumption rate, indicating mitochondrial failure. With increased neuronal maturity, we observed decreased synaptic function in PD iPSC-dopamine neurons, consistent with the requirement for ATP and calcium to support the increase in electrophysiological activity over time. Our work demonstrates that calcium dyshomeostasis and mitochondrial failure impair the higher electrophysiological activity of mature neurons and may underlie the vulnerability of dopamine neurons in PD.
Collapse
Affiliation(s)
- Dayne A. Beccano-Kelly
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX3 7BN, UK
| | - Marta Cherubini
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX3 7BN, UK
| | - Yassine Mousba
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX3 7BN, UK
| | - Kaitlyn M.L. Cramb
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX3 7BN, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - Stefania Giussani
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX3 7BN, UK
| | - Maria Claudia Caiazza
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX3 7BN, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - Pavandeep Rai
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX3 7BN, UK
| | - Siv Vingill
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX3 7BN, UK
| | - Nora Bengoa-Vergniory
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX3 7BN, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - Bryan Ng
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX3 7BN, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - Gabriele Corda
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX3 7BN, UK
| | - Abhirup Banerjee
- Radcliffe Department of Medicine, Division of Cardiovascular Medicine, University of Oxford, Oxford OX3 9DU, UK
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford OX3 7DQ, UK
| | - Jane Vowles
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
- The James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Sally Cowley
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
- The James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Richard Wade-Martins
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX3 7BN, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| |
Collapse
|
15
|
Wang Z, Lin H, Zhang M, Yu W, Zhu C, Wang P, Huang Y, Lv F, Bai H, Wang S. Water-soluble conjugated polymers for bioelectronic systems. MATERIALS HORIZONS 2023; 10:1210-1233. [PMID: 36752220 DOI: 10.1039/d2mh01520j] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Bioelectronics is an interdisciplinary field of research that aims to establish a synergy between electronics and biology. Contributing to a deeper understanding of bioelectronic processes and the built bioelectronic systems, a variety of new phenomena, mechanisms and concepts have been derived in the field of biology, medicine, energy, artificial intelligence science, etc. Organic semiconductors can promote the applications of bioelectronics in improving original performance and creating new features for organisms due to their excellent photoelectric and electrical properties. Recently, water-soluble conjugated polymers (WSCPs) have been employed as a class of ideal interface materials to regulate bioelectronic processes between biological systems and electronic systems, relying on their satisfying ionic conductivity, water-solubility, good biocompatibility and the additional mechanical and electrical properties. In this review, we summarize the prominent contributions of WSCPs in the aspect of the regulation of bioelectronic processes and highlight the latest advances in WSCPs for bioelectronic applications, involving biosynthetic systems, photosynthetic systems, biophotovoltaic systems, and bioelectronic devices. The challenges and outlooks of WSCPs in designing high-performance bioelectronic systems are also discussed.
Collapse
Affiliation(s)
- Zenghao Wang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China.
- College of Chemistry, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Hongrui Lin
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China.
- College of Chemistry, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Miaomiao Zhang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China.
| | - Wen Yu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China.
- College of Chemistry, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Chuanwei Zhu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China.
- College of Chemistry, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Pengcheng Wang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China.
| | - Yiming Huang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China.
| | - Fengting Lv
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China.
| | - Haotian Bai
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China.
| | - Shu Wang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China.
- College of Chemistry, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| |
Collapse
|
16
|
Koshy A, Mathieux E, Stüder F, Bramoulle A, Lieb M, Colombo BM, Gronemeyer H, Mendoza-Parra MA. Synergistic activation of RARβ and RARγ nuclear receptors restores cell specialization during stem cell differentiation by hijacking RARα-controlled programs. Life Sci Alliance 2023; 6:6/2/e202201627. [PMID: 36446525 PMCID: PMC9711859 DOI: 10.26508/lsa.202201627] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 11/10/2022] [Accepted: 11/14/2022] [Indexed: 11/30/2022] Open
Abstract
How cells respond to different external cues to develop along defined cell lineages to form complex tissues is a major question in systems biology. Here, we investigated the potential of retinoic acid receptor (RAR)-selective synthetic agonists to activate the gene regulatory programs driving cell specialization during nervous tissue formation from embryonic carcinoma (P19) and mouse embryonic (E14) stem cells. Specifically, we found that the synergistic activation of the RARβ and RARγ by selective ligands (BMS641 or BMS961) induces cell maturation to specialized neuronal subtypes, and to astrocytes and oligodendrocyte precursors. Using RAR isotype knockout lines exposed to RAR-specific agonists, interrogated by global transcriptome landscaping and in silico modeling of transcription regulatory signal propagation, revealed major RARα-driven gene programs essential for optimal neuronal cell specialization and hijacked by the synergistic activation of the RARβ and RARγ receptors. Overall, this study provides a systems biology view of the gene programs accounting for the previously observed redundancy between RARs, paving the way toward their potential use for directing cell specialization during nervous tissue formation.
Collapse
Affiliation(s)
- Aysis Koshy
- UMR 8030 Génomique Métabolique, Genoscope, Institut François Jacob, CEA, CNRS, University of Evry-val-d'Essonne, University Paris-Saclay, Évry, France
| | - Elodie Mathieux
- UMR 8030 Génomique Métabolique, Genoscope, Institut François Jacob, CEA, CNRS, University of Evry-val-d'Essonne, University Paris-Saclay, Évry, France
| | - François Stüder
- UMR 8030 Génomique Métabolique, Genoscope, Institut François Jacob, CEA, CNRS, University of Evry-val-d'Essonne, University Paris-Saclay, Évry, France
| | - Aude Bramoulle
- UMR 8030 Génomique Métabolique, Genoscope, Institut François Jacob, CEA, CNRS, University of Evry-val-d'Essonne, University Paris-Saclay, Évry, France
| | - Michele Lieb
- Department of Functional Genomics and Cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
| | - Bruno Maria Colombo
- UMR 8030 Génomique Métabolique, Genoscope, Institut François Jacob, CEA, CNRS, University of Evry-val-d'Essonne, University Paris-Saclay, Évry, France
| | - Hinrich Gronemeyer
- Department of Functional Genomics and Cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
| | - Marco Antonio Mendoza-Parra
- UMR 8030 Génomique Métabolique, Genoscope, Institut François Jacob, CEA, CNRS, University of Evry-val-d'Essonne, University Paris-Saclay, Évry, France
| |
Collapse
|
17
|
Arjunan A, Sah DK, Woo M, Song J. Identification of the molecular mechanism of insulin-like growth factor-1 (IGF-1): a promising therapeutic target for neurodegenerative diseases associated with metabolic syndrome. Cell Biosci 2023; 13:16. [PMID: 36691085 PMCID: PMC9872444 DOI: 10.1186/s13578-023-00966-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 01/17/2023] [Indexed: 01/24/2023] Open
Abstract
Neurodegenerative disorders are accompanied by neuronal degeneration and glial dysfunction, resulting in cognitive, psychomotor, and behavioral impairment. Multiple factors including genetic, environmental, metabolic, and oxidant overload contribute to disease progression. Recent evidences suggest that metabolic syndrome is linked to various neurodegenerative diseases. Metabolic syndrome (MetS) is known to be accompanied by symptoms such as hyperglycemia, abdominal obesity, hypertriglyceridemia, and hypertension. Despite advances in knowledge about the pathogenesis of neurodegenerative disorders, effective treatments to combat neurodegenerative disorders caused by MetS have not been developed to date. Insulin growth factor-1 (IGF-1) deficiency has been associated with MetS-related pathologies both in-vivo and in-vitro. IGF-1 is essential for embryonic and adult neurogenesis, neuronal plasticity, neurotropism, angiogenesis, metabolic function, and protein clearance in the brain. Here, we review the evidence for the potential therapeutic effects of IGF-1 in the neurodegeneration related to metabolic syndrome. We elucidate how IGF-1 may be involved in molecular signaling defects that occurs in MetS-related neurodegenerative disorders and highlight the importance of IGF-1 as a potential therapeutic target in MetS-related neurological diseases.
Collapse
Affiliation(s)
- Archana Arjunan
- grid.14005.300000 0001 0356 9399Department of Anatomy, Chonnam National University Medical School, Hwasun, Jeollanam-Do 58128 Republic of Korea
| | - Dhiraj Kumar Sah
- grid.14005.300000 0001 0356 9399Department of Biochemistry, Chonnam National University Medical School, Hwasun, 58128 Republic of Korea ,grid.14005.300000 0001 0356 9399BioMedical Sciences Graduate Program (BMSGP), Chonnam National University, 264 Seoyangro, Hwasun, 58128 Republic of Korea
| | - Minna Woo
- grid.17063.330000 0001 2157 2938Division of Endocrinology and Metabolism, University Health Network and and Banting and Best Diabetes Centre, University of Toronto, Toronto, ON Canada
| | - Juhyun Song
- grid.14005.300000 0001 0356 9399Department of Anatomy, Chonnam National University Medical School, Hwasun, Jeollanam-Do 58128 Republic of Korea ,grid.14005.300000 0001 0356 9399BioMedical Sciences Graduate Program (BMSGP), Chonnam National University, 264 Seoyangro, Hwasun, 58128 Republic of Korea
| |
Collapse
|
18
|
Evidences for Mutant Huntingtin Inducing Musculoskeletal and Brain Growth Impairments via Disturbing Testosterone Biosynthesis in Male Huntington Disease Animals. Cells 2022; 11:cells11233779. [PMID: 36497038 PMCID: PMC9737670 DOI: 10.3390/cells11233779] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/11/2022] [Accepted: 11/20/2022] [Indexed: 11/29/2022] Open
Abstract
Body weight (BW) loss and reduced body mass index (BMI) are the most common peripheral alterations in Huntington disease (HD) and have been found in HD mutation carriers and HD animal models before the manifestation of neurological symptoms. This suggests that, at least in the early disease stage, these changes could be due to abnormal tissue growth rather than tissue atrophy. Moreover, BW and BMI are reported to be more affected in males than females in HD animal models and patients. Here, we confirmed sex-dependent growth alterations in the BACHD rat model for HD and investigated the associated contributing factors. Our results showed growth abnormalities along with decreased plasma testosterone and insulin-like growth factor 1 (IGF-1) levels only in males. Moreover, we demonstrated correlations between growth parameters, IGF-1, and testosterone. Our analyses further revealed an aberrant transcription of testosterone biosynthesis-related genes in the testes of BACHD rats with undisturbed luteinizing hormone (LH)/cAMP/PKA signaling, which plays a key role in regulating the transcription process of some of these genes. In line with the findings in BACHD rats, analyses in the R6/2 mouse model of HD showed similar results. Our findings support the view that mutant huntingtin may induce abnormal growth in males via the dysregulation of gene transcription in the testis, which in turn can affect testosterone biosynthesis.
Collapse
|
19
|
Hédou J, Cederberg KL, Ambati A, Lin L, Farber N, Dauvilliers Y, Quadri M, Bourgin P, Plazzi G, Andlauer O, Hong SC, Huang YS, Leu-Semenescu S, Arnulf I, Taheri S, Mignot E. Proteomic biomarkers of Kleine-Levin syndrome. Sleep 2022; 45:zsac097. [PMID: 35859339 PMCID: PMC9453623 DOI: 10.1093/sleep/zsac097] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 03/21/2022] [Indexed: 07/23/2023] Open
Abstract
STUDY OBJECTIVES Kleine-Levin syndrome (KLS) is characterized by relapsing-remitting episodes of hypersomnia, cognitive impairment, and behavioral disturbances. We quantified cerebrospinal fluid (CSF) and serum proteins in KLS cases and controls. METHODS SomaScan was used to profile 1133 CSF proteins in 30 KLS cases and 134 controls, while 1109 serum proteins were profiled in serum from 26 cases and 65 controls. CSF and serum proteins were both measured in seven cases. Univariate and multivariate analyses were used to find differentially expressed proteins (DEPs). Pathway and tissue enrichment analyses (TEAs) were performed on DEPs. RESULTS Univariate analyses found 28 and 141 proteins differentially expressed in CSF and serum, respectively (false discovery rate <0.1%). Upregulated CSF proteins included IL-34, IL-27, TGF-b, IGF-1, and osteonectin, while DKK4 and vWF were downregulated. Pathway analyses revealed microglial alterations and disrupted blood-brain barrier permeability. Serum profiles show upregulation of Src-family kinases (SFKs), proteins implicated in cellular growth, motility, and activation. TEA analysis of up- and downregulated proteins revealed changes in brain proteins (p < 6 × 10-5), notably from the pons, medulla, and midbrain. A multivariate machine-learning classifier performed robustly, achieving a receiver operating curve area under the curve of 0.90 (95% confidence interval [CI] = 0.78-1.0, p = 0.0006) in CSF and 1.0 (95% CI = 1.0-1.0, p = 0.0002) in serum in validation cohorts, with some commonality across tissues, as the model trained on serum sample also discriminated CSF samples of controls versus KLS cases. CONCLUSIONS Our study identifies proteomic KLS biomarkers with diagnostic potential and provides insight into biological mechanisms that will guide future research in KLS.
Collapse
Affiliation(s)
- Julien Hédou
- Department of Psychiatry and Behavioral Sciences, Center for Sleep Sciences and Medicine, Stanford University, Palo Alto, CA, USA
| | - Katie L Cederberg
- Department of Psychiatry and Behavioral Sciences, Center for Sleep Sciences and Medicine, Stanford University, Palo Alto, CA, USA
| | - Aditya Ambati
- Department of Psychiatry and Behavioral Sciences, Center for Sleep Sciences and Medicine, Stanford University, Palo Alto, CA, USA
| | - Ling Lin
- Department of Psychiatry and Behavioral Sciences, Center for Sleep Sciences and Medicine, Stanford University, Palo Alto, CA, USA
| | - Neal Farber
- Kleine-Levin Syndrome Foundation, Boston, MA, USA
| | - Yves Dauvilliers
- National Reference Centre for Orphan Diseases, Narcolepsy-Rare Hypersomnias, Sleep Unit, Department of Neurology, CHU Montpellier, Univ Montpellier, Montpellier, France
- Department of Neurology, Institute for Neurosciences of Montpellier INM, Univ Montpellier, INSERM, Montpellier, France
| | | | - Patrice Bourgin
- Sleep Disorders Center, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Giuseppe Plazzi
- Department of Biomedical and Neuromotor Sciences, University of Bologna and IRCCS Institute of Neurological Sciences, Bologna, Italy
| | | | - Seung-Chul Hong
- Department of Psychiatry, St. Vincent’s Hospital, Catholic University of Korea, Seoul, South Korea
| | - Yu-Shu Huang
- Department of Child Psychiatry and Sleep Center, Chang Gung Memorial Hospital and University, Taoyuan, Taiwan
| | - Smaranda Leu-Semenescu
- Sleep Disorders, Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris-Sorbonne, National Reference Center for Narcolepsy, Idiopathic Hypersomnia and Kleine-Levin Syndrome, Paris, France
| | - Isabelle Arnulf
- Sleep Disorders, Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris-Sorbonne, National Reference Center for Narcolepsy, Idiopathic Hypersomnia and Kleine-Levin Syndrome, Paris, France
- Sorbonne University, Institut Hospitalo-Universitaire, Institut du Cerveau et de la Moelle, Paris, France
| | - Shahrad Taheri
- Department of Medicine and Clinical Research Core, Weill Cornell Medicine—Qatar, Qatar Foundation—Education City, Doha, Qatar
| | - Emmanuel Mignot
- Corresponding author. Emmanuel Mignot, Center for Narcolepsy and Related Disorders, Stanford University, 3165 Porter Drive, Palo Alto, CA 94305, USA.
| |
Collapse
|
20
|
IGF-1 receptor regulates upward firing rate homeostasis via the mitochondrial calcium uniporter. Proc Natl Acad Sci U S A 2022; 119:e2121040119. [PMID: 35943986 PMCID: PMC9388073 DOI: 10.1073/pnas.2121040119] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
An emerging hypothesis is that neuronal circuits homeostatically maintain a stable spike rate despite continuous environmental changes. This firing rate homeostasis is believed to confer resilience to neurodegeneration and cognitive decline. We show that insulin-like growth factor-1 receptor (IGF-1R) is necessary for homeostatic response of mean firing rate to inactivity, termed “upward firing rate homeostasis.” We show that its mechanism of action is to couple spike bursts with downstream mitochondrial Ca2+ influx via the mitochondrial calcium uniporter complex (MCUc). We propose that MCUc is a homeostatic Ca2+ sensor that triggers the integrated homeostatic response. Firing rate homeostasis may be the principal mechanism by which IGF-1R regulates aging and neurodevelopmental and neurodegenerative disorders. Regulation of firing rate homeostasis constitutes a fundamental property of central neural circuits. While intracellular Ca2+ has long been hypothesized to be a feedback control signal, the molecular machinery enabling a network-wide homeostatic response remains largely unknown. We show that deletion of insulin-like growth factor-1 receptor (IGF-1R) limits firing rate homeostasis in response to inactivity, without altering the distribution of baseline firing rates. The deficient firing rate homeostatic response was due to disruption of both postsynaptic and intrinsic plasticity. At the cellular level, we detected a fraction of IGF-1Rs in mitochondria, colocalized with the mitochondrial calcium uniporter complex (MCUc). IGF-1R deletion suppressed transcription of the MCUc members and burst-evoked mitochondrial Ca2+ (mitoCa2+) by weakening mitochondria-to-cytosol Ca2+ coupling. Overexpression of either mitochondria-targeted IGF-1R or MCUc in IGF-1R–deficient neurons was sufficient to rescue the deficits in burst-to-mitoCa2+ coupling and firing rate homeostasis. Our findings indicate that mitochondrial IGF-1R is a key regulator of the integrated homeostatic response by tuning the reliability of burst transfer by MCUc. Based on these results, we propose that MCUc acts as a homeostatic Ca2+ sensor. Faulty activation of MCUc may drive dysregulation of firing rate homeostasis in aging and in brain disorders associated with aberrant IGF-1R/MCUc signaling.
Collapse
|
21
|
Leung HW, Foo G, VanDongen A. Arc Regulates Transcription of Genes for Plasticity, Excitability and Alzheimer’s Disease. Biomedicines 2022; 10:biomedicines10081946. [PMID: 36009494 PMCID: PMC9405677 DOI: 10.3390/biomedicines10081946] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/21/2022] [Accepted: 07/25/2022] [Indexed: 02/06/2023] Open
Abstract
The immediate early gene Arc is a master regulator of synaptic function and a critical determinant of memory consolidation. Here, we show that Arc interacts with dynamic chromatin and closely associates with histone markers for active enhancers and transcription in cultured rat hippocampal neurons. Both these histone modifications, H3K27Ac and H3K9Ac, have recently been shown to be upregulated in late-onset Alzheimer’s disease (AD). When Arc induction by pharmacological network activation was prevented using a short hairpin RNA, the expression profile was altered for over 1900 genes, which included genes associated with synaptic function, neuronal plasticity, intrinsic excitability, and signalling pathways. Interestingly, about 100 Arc-dependent genes are associated with the pathophysiology of AD. When endogenous Arc expression was induced in HEK293T cells, the transcription of many neuronal genes was increased, suggesting that Arc can control expression in the absence of activated signalling pathways. Taken together, these data establish Arc as a master regulator of neuronal activity-dependent gene expression and suggest that it plays a significant role in the pathophysiology of AD.
Collapse
Affiliation(s)
| | - Gabriel Foo
- Duke-NUS Medical School, Singapore 169857, Singapore
| | - Antonius VanDongen
- Duke-NUS Medical School, Singapore 169857, Singapore
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
- Correspondence:
| |
Collapse
|
22
|
Insulin-like growth factor 1 regulates excitatory synaptic transmission in pyramidal neurons from adult prefrontal cortex. Neuropharmacology 2022; 217:109204. [PMID: 35931212 DOI: 10.1016/j.neuropharm.2022.109204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 07/07/2022] [Accepted: 07/25/2022] [Indexed: 11/24/2022]
Abstract
Insulin-like growth factor 1 (IGF1) influences synaptic function in addition to its role in brain development and aging. Although the expression levels of IGF1 and IGF1 receptor (IGF1R) peak during development and decline with age, the adult brain has abundant IGF1 or IGF1R expression. Studies reveal that IGF1 regulates the synaptic transmission in neurons from young animals. However, the action of IGF1 on neurons in the adult brain is still unclear. Here, we used prefrontal cortical (PFC) slices from adult mice (∼8 weeks old) to characterize the role of IGF1 on excitatory synaptic transmission in pyramidal neurons and the underlying molecular mechanisms. We first validated IGF1R expression in pyramidal neurons using translating ribosomal affinity purification assay. Then, using whole-cell patch-clamp recording, we found that IGF1 attenuated the amplitude of evoked excitatory postsynaptic current (EPSC) without affecting the frequency and amplitude of miniature EPSC. Furthermore, this decrease in excitatory neurotransmission was blocked by pharmacological inhibition of IGF1R or conditionally knockdown of IGF1R in PFC pyramidal neurons. In addition, we determined that IGF1-induced decrease of EPSC amplitude was due to postsynaptic effect (internalization of a-amino-3-hydroxy-5-methyl-4- isoxazolepropionic acid receptors [AMPAR]) rather than presynaptic glutamate release. Finally, we found that inhibition of metabotropic glutamate receptor subtype-1 (mGluR1) abolished IGF1-induced attenuation of evoked EPSC amplitude and decrease of AMPAR expression at synaptic membrane, suggesting mGluR1-mediated endocytosis of AMPAR was involved. Taken together, these data provide the first evidence that IGF1 regulates excitatory synaptic transmission in adult PFC via the interaction between IGF1R-dependent signaling pathway and mGluR1-mediated AMPAR endocytosis.
Collapse
|
23
|
Kelly EA, Contreras J, Duan A, Vassell R, Fudge JL. Unbiased Stereological Estimates of Dopaminergic and GABAergic Neurons in the A10, A9, and A8 Subregions in the Young Male Macaque. Neuroscience 2022; 496:152-164. [PMID: 35738547 PMCID: PMC9329254 DOI: 10.1016/j.neuroscience.2022.06.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 05/25/2022] [Accepted: 06/10/2022] [Indexed: 11/28/2022]
Abstract
The ventral midbrain is the primary source of dopamine- (DA) expressing neurons in most species. GABA-ergic and glutamatergic cell populations are intermixed among DA-expressing cells and purported to regulate both local and long-range dopamine neuron activity. Most work has been conducted in rodent models, however due to evolutionary expansion of the ventral midbrain in primates, the increased size and complexity of DA subpopulations warrants further investigation. Here, we quantified the number of DA neurons, and their GABA-ergic complement in classic DA cell groups A10 (midline ventral tegmental area nuclei [VTA] and parabrachial pigmented nucleus [PBP]), A9 (substantia nigra, pars compacta [SNc]) and A8 (retrorubral field [RRF]) in the macaque. Because the PBP is a disproportionately expanded feature of the A10 group, and has unique connectional features in monkeys, we analyzed A10 data by dividing it into 'classic' midline nuclei and the PBP. Unbiased stereology revealed total putative DA neuron counts to be 210,238 ± 17,127 (A10 = 110,319 ± 9649, A9 = 87,399 ± 7751 and A8 = 12,520 ± 827). Putative GABAergic neurons were fewer overall, and evenly dispersed across the DA subpopulations (GAD67 = 71,215 ± 5663; A10 = 16,836 ± 2743; A9 = 24,855 ± 3144 and A8 = 12,633 ± 3557). Calculating the GAD67/TH ratio for each subregion revealed differential balances of these two cell types across the DA subregions. The A8 subregion had the highest complement of GAD67-positive neurons compared to TH-positive neurons (1:1), suggesting a potentially high capacity for GABAergic inhibition of DA output in this region.
Collapse
Affiliation(s)
- Emily A Kelly
- Department of Neuroscience, University of Rochester Medical Center, United States
| | - Jancy Contreras
- Department of Neuroscience, The City University of New York, United States
| | - Annie Duan
- Department of Neuroscience, University of Rochester Medical Center, United States
| | - Rochelle Vassell
- Department of Neuroscience, University of Rochester Medical Center, United States
| | - Julie L Fudge
- Department of Neuroscience, University of Rochester Medical Center, United States; Department of Psychiatry, University of Rochester Medical Center, United States.
| |
Collapse
|
24
|
IGF-1 release in the medial prefrontal cortex mediates the rapid and sustained antidepressant-like actions of ketamine. Transl Psychiatry 2022; 12:178. [PMID: 35577782 PMCID: PMC9110717 DOI: 10.1038/s41398-022-01943-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 04/15/2022] [Accepted: 04/20/2022] [Indexed: 12/13/2022] Open
Abstract
Ketamine, an N-methyl-D-aspartate receptor antagonist, exerts rapid and sustained antidepressant actions. Preclinical studies demonstrated that the release of brain-derived neurotrophic factor (BDNF) and vascular endothelial growth factor in the medial prefrontal cortex (mPFC) is essential for the antidepressant-like effects of ketamine. However, the role of other neurotrophic factors in the antidepressant-like effects of ketamine has not been fully investigated. Since the intra-mPFC infusion of insulin-like growth factor 1 (IGF-1) reportedly produced antidepressant-like effects, the present study examined the role of endogenous intra-mPFC IGF-1 signaling in the antidepressant-like actions of ketamine. In vivo microdialysis showed that ketamine (10 and 30 mg/kg) significantly increased extracellular IGF-1 levels in the mPFC of male C57BL/6J mice for at least 5 h. Infusion of an IGF-1 neutralizing antibody (nAb; 160 ng/side) into the mPFC 15 min before or 2 h after ketamine injection blocked the antidepressant-like effects of ketamine in three different behavioral paradigms (forced swim, female urine sniffing, and novelty-suppressed feeding tests were conducted 1, 3 and 4 days post-ketamine, respectively). The ketamine-like antidepressant-like actions of the intra-mPFC infusion of BDNF (100 ng/side) and IGF-1 (50 ng/side) respectively were not blocked by co-infused IGF-1 nAb and BDNF nAb (200 ng/side). Moreover, intra-mPFC infusion of IGF-1 nAb 2 h post-ketamine blocked the antidepressant-like effects of ketamine in a murine lipopolysaccharide (LPS)-induced depression model. Intra-mPFC IGF-1 infusion also produced antidepressant-like effects in the LPS-challenged mice via mechanistic target of rapamycin complex 1 activation. These results suggest that persistent release of IGF-1, independently of BDNF, in the mPFC is essential for the antidepressant-like actions of ketamine.
Collapse
|
25
|
Noriega-Prieto JA, Maglio LE, Ibáñez-Santana S, de Sevilla DF. Endocannabinoid and Nitric Oxide-Dependent IGF-I-Mediated Synaptic Plasticity at Mice Barrel Cortex. Cells 2022; 11:cells11101641. [PMID: 35626678 PMCID: PMC9140009 DOI: 10.3390/cells11101641] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 04/22/2022] [Accepted: 05/12/2022] [Indexed: 11/28/2022] Open
Abstract
Insulin-like growth factor-I (IGF-I) signaling plays a key role in learning and memory. IGF-I increases the spiking and induces synaptic plasticity in the mice barrel cortex (Noriega-Prieto et al., 2021), favoring the induction of the long-term potentiation (LTP) by Spike Timing-Dependent Protocols (STDP) (Noriega-Prieto et al., 2021). Here, we studied whether these IGF-I effects depend on endocannabinoids (eCBs) and nitric oxide (NO). We recorded both excitatory postsynaptic currents (EPSCs) and inhibitory postsynaptic currents (IPSCs) evoked by stimulation of the basal dendrites of layer II/III pyramidal neurons of the Barrel Cortex and analyzed the effect of IGF-I in the presence of a CB1R antagonist, AM251, and inhibitor of the NO synthesis, L-NAME, to prevent the eCBs and the NO-mediated signaling. Interestingly, L-NAME abolished any modulatory effect of the IGF-I-induced excitatory and inhibitory transmission changes, suggesting the essential role of NO. Surprisingly, the inhibition of CB1Rs did not only block the potentiation of EPSCs but reversed to a depression, highlighting the remarkable functions of the eCB system. In conclusion, eCBs and NO play a vital role in deciding the sign of the effects induced by IGF-I in the neocortex, suggesting a neuromodulatory interplay among IGF-I, NO, and eCBs.
Collapse
Affiliation(s)
- José Antonio Noriega-Prieto
- Departamento de Anatomía, Histología y Neurociencia, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (J.A.N.-P.); (L.E.M.); (S.I.-S.)
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Laura Eva Maglio
- Departamento de Anatomía, Histología y Neurociencia, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (J.A.N.-P.); (L.E.M.); (S.I.-S.)
| | - Sara Ibáñez-Santana
- Departamento de Anatomía, Histología y Neurociencia, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (J.A.N.-P.); (L.E.M.); (S.I.-S.)
| | - David Fernández de Sevilla
- Departamento de Anatomía, Histología y Neurociencia, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (J.A.N.-P.); (L.E.M.); (S.I.-S.)
- Correspondence:
| |
Collapse
|
26
|
Upreti S, Sen S, Nag TC, Ghosh MP. Insulin like growth factor-1 works synergistically with dopamine to attenuate diabetic retinopathy by downregulating vascular endothelial growth factor. Biomed Pharmacother 2022; 149:112868. [PMID: 35378500 DOI: 10.1016/j.biopha.2022.112868] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 03/16/2022] [Accepted: 03/23/2022] [Indexed: 11/02/2022] Open
Abstract
AIM Levels of Insulin-like growth factor-1 (IGF-1), a proangiogenic growth factor is elevated and dopamine downregulated in proliferative diabetic retinopathy (PDR). This study aims to investigate whether IGF-1 with dopamine can together modulate vascular endothelial growth factor (VEGF) to prevent proliferative diabetic retinopathy while also attenuating angiogenic effects of IGF-1. METHODS Effect of combination of levodopa L-Dopa with IGF-1 was tested on normal retinal pigment epithelium cells (ARPE-19) and human umbilical vein endothelial cells (HUVEC), followed by tube formation. Invivo analysis of anti-angiogenic potential assessed by chick chorioallantoic membrane (CAM) assay. Diabetes induction in wistar rats at two time points, 12 and 16 weeks, treated with L-Dopa+IGF-1 and analysed for morphological variations, serum and tissue dopamine levels, gene expression by real-time PCR and western blot assay. RESULTS L-Dopa+IGF-1 on ARPE-19 cells caused no toxicity and worked synergistically. Reduced number of vessels observed. Significant improvement in inner retina thickness (*p < 0.05) was observed when L-Dopa was given alone and/or with IGF-1. Dopamine levels improved significantly in both serum and tissue (*p < 0.05). Levels of VEGF and IGF-1 receptors reduced significantly in 12 weeks. Western studies suggest that L-Dopa+IGF-1 modulates its effects via Akt/ERK dependent pathway. CONCLUSION First ever report on synergistic effect of L-Dopa+IGF-1 in a rat model of diabetic retinopathy. Even though the effect of L-Dopa in combination with IGF-1 is comparable to levels of L-Dopa alone, this study presents an interesting finding of neuroprotective function of IGF-1, which has been studied in disease models of Parkinson's but not diabetes.
Collapse
Affiliation(s)
- Shikha Upreti
- Ocular Pharmacology and Therapeutics Lab, Centre for Medical Biotechnology, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida 201313, India.
| | - Seema Sen
- Department of Ocular Pathology, Dr R.P. Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India.
| | - Tapas Chandra Nag
- Department of Anatomy, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India.
| | - Madhumita P Ghosh
- Ocular Pharmacology and Therapeutics Lab, Centre for Medical Biotechnology, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida 201313, India.
| |
Collapse
|
27
|
Kohno K, Shirasaka R, Yoshihara K, Mikuriya S, Tanaka K, Takanami K, Inoue K, Sakamoto H, Ohkawa Y, Masuda T, Tsuda M. A spinal microglia population involved in remitting and relapsing neuropathic pain. Science 2022; 376:86-90. [PMID: 35357926 DOI: 10.1126/science.abf6805] [Citation(s) in RCA: 128] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Neuropathic pain is often caused by injury and diseases that affect the somatosensory system. Although pain development has been well studied, pain recovery mechanisms remain largely unknown. Here, we found that CD11c-expressing spinal microglia appear after the development of behavioral pain hypersensitivity following nerve injury. Nerve-injured mice with spinal CD11c+ microglial depletion failed to recover spontaneously from this hypersensitivity. CD11c+ microglia expressed insulin-like growth factor-1 (IGF1), and interference with IGF1 signaling recapitulated the impairment in pain recovery. In pain-recovered mice, the depletion of CD11c+ microglia or the interruption of IGF1 signaling resulted in a relapse in pain hypersensitivity. Our findings reveal a mechanism for the remission and recurrence of neuropathic pain, providing potential targets for therapeutic strategies.
Collapse
Affiliation(s)
- Keita Kohno
- Department of Life Innovation, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan.,Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Ryoji Shirasaka
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Kohei Yoshihara
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Satsuki Mikuriya
- Department of Life Innovation, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Kaori Tanaka
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Keiko Takanami
- Ushimado Marine Institute, Graduate School of Natural Science and Technology, Okayama University, Setouchi, Japan.,Mouse Genomics Resources Laboratory, National Institute of Genetics, Shizuoka, Japan
| | - Kazuhide Inoue
- Kyushu University Institute for Advanced Study, Fukuoka, Japan
| | - Hirotaka Sakamoto
- Ushimado Marine Institute, Graduate School of Natural Science and Technology, Okayama University, Setouchi, Japan
| | - Yasuyuki Ohkawa
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Takahiro Masuda
- Department of Life Innovation, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan.,Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Makoto Tsuda
- Department of Life Innovation, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan.,Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
28
|
IGF-1 as a Potential Therapy for Spinocerebellar Ataxia Type 3. Biomedicines 2022; 10:biomedicines10020505. [PMID: 35203722 PMCID: PMC8962315 DOI: 10.3390/biomedicines10020505] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/18/2022] [Accepted: 02/19/2022] [Indexed: 02/04/2023] Open
Abstract
Although the effects of growth hormone (GH) therapy on spinocerebellar ataxia type 3 (SCA3) have been examined in transgenic SCA3 mice, it still poses a nonnegligible risk of cancer when used for a long term. This study investigated the efficacy of IGF-1, a downstream mediator of GH, in vivo for SCA3 treatment. IGF-1 (50 mg/kg) or saline, once a week, was intraperitoneally injected to SCA3 84Q transgenic mice harboring a human ATXN3 gene with a pathogenic expanded 84 cytosine–adenine–guanine (CAG) repeat motif at 9 months of age. Compared with the control mice harboring a 15 CAG repeat motif, the SCA3 84Q mice treated with IGF-1 for 9 months exhibited the improvement only in locomotor function and minimized degeneration of the cerebellar cortex as indicated by the survival of more Purkinje cells with a more favorable mitochondrial function along with a decrease in oxidative stress caused by DNA damage. These findings could be attributable to the inhibition of mitochondrial fission, resulting in mitochondrial fusion, and decreased immunofluorescence staining in aggresome formation and ataxin-3 mutant protein levels, possibly through the enhancement of autophagy. The findings of this study show the therapeutic potential effect of IGF-1 injection for SCA3 to prevent the exacerbation of disease progress.
Collapse
|
29
|
Tomioka M, Jang MS, Iino Y. DAF-2c signaling promotes taste avoidance after starvation in Caenorhabditis elegans by controlling distinct phospholipase C isozymes. Commun Biol 2022; 5:30. [PMID: 35017611 PMCID: PMC8752840 DOI: 10.1038/s42003-021-02956-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 12/09/2021] [Indexed: 12/19/2022] Open
Abstract
Previously, we reported that DAF-2c, an axonal insulin receptor isoform in Caenorhabditis elegans, acts in the ASER gustatory neuron to regulate taste avoidance learning, a process in which worms learn to avoid salt concentrations experienced during starvation. Here, we show that secretion of INS-1, an insulin-like peptide, after starvation conditioning is sufficient to drive taste avoidance via DAF-2c signaling. Starvation conditioning enhances the salt-triggered activity of AIA neurons, the main sites of INS-1 release, which potentially promotes feedback signaling to ASER to maintain DAF-2c activity during taste avoidance. Genetic studies suggest that DAF-2c-Akt signaling promotes high-salt avoidance via a decrease in PLCβ activity. On the other hand, the DAF-2c pathway promotes low-salt avoidance via PLCε and putative Akt phosphorylation sites on PLCε are essential for taste avoidance. Our findings imply that animals disperse from the location at which they experience starvation by controlling distinct PLC isozymes via DAF-2c.
Collapse
Affiliation(s)
- Masahiro Tomioka
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| | - Moon Sun Jang
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
- Neuroscience Institute, Graduate School of Science, Nagoya University, Nagoya, Aichi, 464-8602, Japan
| | - Yuichi Iino
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| |
Collapse
|
30
|
Sallam NA, Borgland SL. Insulin and endocannabinoids in the mesolimbic system. J Neuroendocrinol 2021; 33:e12965. [PMID: 33856071 DOI: 10.1111/jne.12965] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 02/19/2021] [Accepted: 03/08/2021] [Indexed: 12/31/2022]
Abstract
Easy access to palatable food and an abundance of food-related cues exacerbate non-homeostatic feeding. The metabolic and economical sequelae of non-homeostatic feeding outweigh those of homeostatic feeding and contribute significantly to the global obesity pandemic. The mesolimbic dopamine system is the primary central circuit that governs the motivation to consume food. Insulin and endocannabinoids (eCBs) are two major, presumably opposing, players in regulating homeostatic and non-homeostatic feeding centrally and peripherally. Insulin is generally regarded as a postprandial satiety signal, whereas eCBs mainly function as pre-prandial orexinergic signals. In this review, we discuss the effects of insulin and eCB-mediated actions within the mesolimbic pathways. We propose that insulin and eCBs have regional- and time course-dependent roles. We discuss their mechanisms of actions in the ventral tegmental area and nucleus accumbens, as well as how their mechanisms converge to finely tune dopaminergic activity and food intake.
Collapse
Affiliation(s)
- Nada A Sallam
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Stephanie L Borgland
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
31
|
López‐Gambero AJ, Rodríguez de Fonseca F, Suárez J. Energy sensors in drug addiction: A potential therapeutic target. Addict Biol 2021; 26:e12936. [PMID: 32638485 DOI: 10.1111/adb.12936] [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: 02/22/2020] [Revised: 06/10/2020] [Accepted: 06/15/2020] [Indexed: 01/05/2023]
Abstract
Addiction is defined as the repeated exposure and compulsive seek of psychotropic drugs that, despite the harmful effects, generate relapse after the abstinence period. The psychophysiological processes associated with drug addiction (acquisition/expression, withdrawal, and relapse) imply important alterations in neurotransmission and changes in presynaptic and postsynaptic plasticity and cellular structure (neuroadaptations) in neurons of the reward circuits (dopaminergic neuronal activity) and other corticolimbic regions. These neuroadaptation mechanisms imply important changes in neuronal energy balance and protein synthesis machinery. Scientific literature links drug-induced stimulation of dopaminergic and glutamatergic pathways along with presence of neurotrophic factors with alterations in synaptic plasticity and membrane excitability driven by metabolic sensors. Here, we provide current knowledge of the role of molecular targets that constitute true metabolic/energy sensors such as AMPK, mTOR, ERK, or KATP in the development of the different phases of addiction standing out the main brain regions (ventral tegmental area, nucleus accumbens, hippocampus, and amygdala) constituting the hubs in the development of addiction. Because the available treatments show very limited effectiveness, evaluating the drug efficacy of AMPK and mTOR specific modulators opens up the possibility of testing novel pharmacotherapies for an individualized approach in drug abuse.
Collapse
Affiliation(s)
- Antonio Jesús López‐Gambero
- Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga Universidad de Málaga Málaga Spain
| | - Fernando Rodríguez de Fonseca
- Instituto de Investigación Biomédica de Málaga (IBIMA), UGC Salud Mental Hospital Regional Universitario de Málaga Málaga Spain
| | - Juan Suárez
- Instituto de Investigación Biomédica de Málaga (IBIMA), UGC Salud Mental Hospital Regional Universitario de Málaga Málaga Spain
| |
Collapse
|
32
|
Matsuno H, Tsuchimine S, Fukuzato N, O'Hashi K, Kunugi H, Sohya K. Sirtuin 6 is a regulator of dendrite morphogenesis in rat hippocampal neurons. Neurochem Int 2021; 145:104959. [PMID: 33444676 DOI: 10.1016/j.neuint.2021.104959] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 12/15/2020] [Accepted: 01/05/2021] [Indexed: 01/14/2023]
Abstract
Sirtuin 6 (SIRT6), a member of the Sirtuin family, acts as nicotinamide adenine dinucleotide (NAD)-dependent protein deacetylase, mono-adenosine diphosphate (ADP)-ribosyltransferase, and fatty acid deacylase, and plays critical roles in inflammation, aging, glycolysis, and DNA repair. Accumulating evidence has suggested that SIRT6 is involved in brain functions such as neuronal differentiation, neurogenesis, and learning and memory. However, the precise molecular roles of SIRT6 during neuronal circuit formation are not yet well understood. In this study, we tried to elucidate molecular roles of SIRT6 on neurite development by using primary-cultured hippocampal neurons. We observed that SIRT6 was abundantly localized in the nucleus, and its expression was markedly increased during neurite outgrowth and synaptogenesis. By using shRNA-mediated SIRT6-knockdown, we show that both dendritic length and the number of dendrite branches were significantly reduced in the SIRT6-knockdown neurons. Microarray and subsequent gene ontology analysis revealed that reducing SIRT6 caused the downregulation of immediate early genes (IEGs) and alteration of several biological processes including MAPK (ERK1/2) signaling. We found that nuclear accumulation of phosphorylated ERK1/2 was significantly reduced in SIRT6-knockdown neurons. Overexpression of SIRT6 promoted dendritic length and branching, but the mutants lacking deacetylase activity had no significant effect on the dendritic morphology. Collectively, the presented findings reveal a role of SIRT6 in dendrite morphogenesis, and suggest that SIRT6 may act as an important regulator of ERK1/2 signaling pathway that mediates IEG expression, which leads to dendritic development.
Collapse
Affiliation(s)
- Hitomi Matsuno
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), 4-1-1 Ogawa-Higashi, Kodaira, Tokyo, 187-8502, Japan.
| | - Shoko Tsuchimine
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), 4-1-1 Ogawa-Higashi, Kodaira, Tokyo, 187-8502, Japan
| | - Noriko Fukuzato
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), 4-1-1 Ogawa-Higashi, Kodaira, Tokyo, 187-8502, Japan
| | - Kazunori O'Hashi
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), 4-1-1 Ogawa-Higashi, Kodaira, Tokyo, 187-8502, Japan; Department of Pharmacology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Hiroshi Kunugi
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), 4-1-1 Ogawa-Higashi, Kodaira, Tokyo, 187-8502, Japan; Department of Psychiatry, Teikyo University School of Medicine, 2-11-1, Kaga, Itabashi-ku, Tokyo, 173-8605, Japan
| | - Kazuhiro Sohya
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), 4-1-1 Ogawa-Higashi, Kodaira, Tokyo, 187-8502, Japan.
| |
Collapse
|
33
|
Guerra-Cantera S, Frago LM, Collado-Pérez R, Canelles S, Ros P, Freire-Regatillo A, Jiménez-Hernaiz M, Barrios V, Argente J, Chowen JA. Sex Differences in Metabolic Recuperation After Weight Loss in High Fat Diet-Induced Obese Mice. Front Endocrinol (Lausanne) 2021; 12:796661. [PMID: 34975768 PMCID: PMC8716724 DOI: 10.3389/fendo.2021.796661] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 11/16/2021] [Indexed: 01/10/2023] Open
Abstract
Dietary intervention is a common tactic employed to curtail the current obesity epidemic. Changes in nutritional status alter metabolic hormones such as insulin or leptin, as well as the insulin-like growth factor (IGF) system, but little is known about restoration of these parameters after weight loss in obese subjects and if this differs between the sexes, especially regarding the IGF system. Here male and female mice received a high fat diet (HFD) or chow for 8 weeks, then half of the HFD mice were changed to chow (HFDCH) for 4 weeks. Both sexes gained weight (p < 0.001) and increased their energy intake (p < 0.001) and basal glycemia (p < 0.5) on the HFD, with these parameters normalizing after switching to chow but at different rates in males and females. In both sexes HFD decreased hypothalamic NPY and AgRP (p < 0.001) and increased POMC (p < 0.001) mRNA levels, with all normalizing in HFDCH mice, whereas the HFD-induced decrease in ObR did not normalize (p < 0.05). All HFD mice had abnormal glucose tolerance tests (p < 0.001), with males clearly more affected, that normalized when returned to chow. HFD increased insulin levels and HOMA index (p < 0.01) in both sexes, but only HFDCH males normalized this parameter. Returning to chow normalized the HFD-induced increase in circulating leptin (p < 0.001), total IGF1 (p < 0.001), IGF2 (p < 0.001, only in females) and IGFBP3 (p < 0.001), whereas free IGF1 levels remained elevated (p < 0.01). In males IGFBP2 decreased with HFD and normalized with chow (p < 0.001), with no changes in females. Although returning to a healthy diet improved of most metabolic parameters analyzed, fIGF1 levels remained elevated and hypothalamic ObR decreased in both sexes. Moreover, there was sex differences in both the response to HFD and the switch to chow including circulating levels of IGF2 and IGFBP2, factors previously reported to be involved in glucose metabolism. Indeed, glucose metabolism was also differentially modified in males and females, suggesting that these observations could be related.
Collapse
Affiliation(s)
- Santiago Guerra-Cantera
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Laura M. Frago
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Roberto Collado-Pérez
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
| | - Sandra Canelles
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Purificación Ros
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Department of Pediatrics, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Alejandra Freire-Regatillo
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - María Jiménez-Hernaiz
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Vicente Barrios
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Jesús Argente
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- IMDEA Food Institute, CEI UAM + CSIC, Madrid, Spain
- *Correspondence: Julie A. Chowen, ; Jesús Argente,
| | - Julie A. Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- IMDEA Food Institute, CEI UAM + CSIC, Madrid, Spain
- *Correspondence: Julie A. Chowen, ; Jesús Argente,
| |
Collapse
|
34
|
Javed S, Selliah T, Lee YJ, Huang WH. Dosage-sensitive genes in autism spectrum disorders: From neurobiology to therapy. Neurosci Biobehav Rev 2020; 118:538-567. [PMID: 32858083 DOI: 10.1016/j.neubiorev.2020.08.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/26/2020] [Accepted: 08/17/2020] [Indexed: 12/24/2022]
Abstract
Autism spectrum disorders (ASDs) are a group of heterogenous neurodevelopmental disorders affecting 1 in 59 children. Syndromic ASDs are commonly associated with chromosomal rearrangements or dosage imbalance involving a single gene. Many of these genes are dosage-sensitive and regulate transcription, protein homeostasis, and synaptic function in the brain. Despite vastly different molecular perturbations, syndromic ASDs share core symptoms including social dysfunction and repetitive behavior. However, each ASD subtype has a unique pathogenic mechanism and combination of comorbidities that require individual attention. We have learned a great deal about how these dosage-sensitive genes control brain development and behaviors from genetically-engineered mice. Here we describe the clinical features of eight monogenic neurodevelopmental disorders caused by dosage imbalance of four genes, as well as recent advances in using genetic mouse models to understand their pathogenic mechanisms and develop intervention strategies. We propose that applying newly developed quantitative molecular and neuroscience technologies will advance our understanding of the unique neurobiology of each disorder and enable the development of personalized therapy.
Collapse
Affiliation(s)
- Sehrish Javed
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, The Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Tharushan Selliah
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, The Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Yu-Ju Lee
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, The Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Wei-Hsiang Huang
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, The Research Institute of the McGill University Health Centre, Montréal, Québec, Canada.
| |
Collapse
|
35
|
Sleep and the GH/IGF-1 axis: Consequences and countermeasures of sleep loss/disorders. Sleep Med Rev 2019; 49:101223. [PMID: 31778943 DOI: 10.1016/j.smrv.2019.101223] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 10/21/2019] [Accepted: 10/21/2019] [Indexed: 12/15/2022]
Abstract
This article presents an up-to-date review of the state-of-the-art knowledge regarding the effect of sleep on the anabolic growth hormone/insulin-like growth factor-1 (GH/IGF-1) axis. This axis is involved in learning and memory and neuroprotection at the central level, and in the crosstalk between sleep and the immune system, with respect to its anti-inflammatory properties. We also aim to provide insight into the consequences of sleep loss on cognitive capacities in healthy individuals and patients with obstructive sleep apnea (OSA), regarding the mechanistic association with the GH/IGF-1 axis. Finally, this review examines the inflammatory/endocrine pathways that are affected by sleep loss, and which may consequently interact with the GH/IGF-1 axis. The deleterious effects of sleep loss include fatigue, and can cause several adverse age-dependent health outcomes. It is therefore important to improve our understanding of the fundamental physiology underlying these effects in order to better apply non-pharmacological countermeasures (e.g., sleep strategies, exercise training, continuous positive airway pressure therapy) as well as pharmacological solutions, so as to limit the deleterious consequences of sleep loss/disorders.
Collapse
|
36
|
Tian Y, Song M. Sevoflurane Affects Memory Through Impairing Insulin-Like Growth Factor Receptor Signaling. J Alzheimers Dis 2019; 71:825-832. [PMID: 31450504 DOI: 10.3233/jad-190596] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Yuan Tian
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingrui Song
- Department of Neurology, Shanghai Changzheng Hospital, the Second Military Medical University, Shanghai, China
| |
Collapse
|
37
|
Huang X, Su C, Chen S, Teng J, Zhao L, Chen S. Effect of klotho as an ageing suppressor on insulin signalling cascade in brain of d-galactose mice by Yisui moxibustion. BIOTECHNOL BIOTEC EQ 2019. [DOI: 10.1080/13102818.2019.1665476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Affiliation(s)
- Xinge Huang
- Department of Acupuncture and Moxibustion, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, PR China
| | - Chuanli Su
- Department of Pathology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, PR China
| | - Shuyan Chen
- Department of Acupuncture and Moxibustion Affiliated Orthopedics Surgery, Jianxiang Hospital, Foshan, PR China
| | - Jinlong Teng
- Acupuncture and Massage College, Guangxi University of Chinese Medicine, Nanning, PR China
| | - Lihua Zhao
- Department of Acupuncture and Moxibustion, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, PR China
| | - Shangjie Chen
- Department of Rehabilitation, Bao’an Hospital, Southern Medical University, Shenzhen, PR China
| |
Collapse
|
38
|
Di Liegro CM, Schiera G, Proia P, Di Liegro I. Physical Activity and Brain Health. Genes (Basel) 2019; 10:genes10090720. [PMID: 31533339 PMCID: PMC6770965 DOI: 10.3390/genes10090720] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 09/12/2019] [Indexed: 12/16/2022] Open
Abstract
Physical activity (PA) has been central in the life of our species for most of its history, and thus shaped our physiology during evolution. However, only recently the health consequences of a sedentary lifestyle, and of highly energetic diets, are becoming clear. It has been also acknowledged that lifestyle and diet can induce epigenetic modifications which modify chromatin structure and gene expression, thus causing even heritable metabolic outcomes. Many studies have shown that PA can reverse at least some of the unwanted effects of sedentary lifestyle, and can also contribute in delaying brain aging and degenerative pathologies such as Alzheimer’s Disease, diabetes, and multiple sclerosis. Most importantly, PA improves cognitive processes and memory, has analgesic and antidepressant effects, and even induces a sense of wellbeing, giving strength to the ancient principle of “mens sana in corpore sano” (i.e., a sound mind in a sound body). In this review we will discuss the potential mechanisms underlying the effects of PA on brain health, focusing on hormones, neurotrophins, and neurotransmitters, the release of which is modulated by PA, as well as on the intra- and extra-cellular pathways that regulate the expression of some of the genes involved.
Collapse
Affiliation(s)
- Carlo Maria Di Liegro
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche) (STEBICEF), University of Palermo, 90128 Palermo, Italy.
| | - Gabriella Schiera
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche) (STEBICEF), University of Palermo, 90128 Palermo, Italy.
| | - Patrizia Proia
- Department of Psychology, Educational Science and Human Movement (Dipartimento di Scienze Psicologiche, Pedagogiche, dell'Esercizio fisico e della Formazione), University of Palermo, 90128 Palermo, Italy.
| | - Italia Di Liegro
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (Dipartimento di Biomedicina, Neuroscienze e Diagnostica avanzata) (Bi.N.D.), University of Palermo, 90127 Palermo, Italy.
| |
Collapse
|