1
|
Rastogi M, Bartolucci M, Nanni M, Aloisio M, Vozzi D, Petretto A, Contestabile A, Cancedda L. Integrative multi-omic analysis reveals conserved cell-projection deficits in human Down syndrome brains. Neuron 2024; 112:2503-2523.e10. [PMID: 38810652 DOI: 10.1016/j.neuron.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 03/17/2024] [Accepted: 05/01/2024] [Indexed: 05/31/2024]
Abstract
Down syndrome (DS) is the most common genetic cause of cognitive disability. However, it is largely unclear how triplication of a small gene subset may impinge on diverse aspects of DS brain physiopathology. Here, we took a multi-omic approach and simultaneously analyzed by RNA-seq and proteomics the expression signatures of two diverse regions of human postmortem DS brains. We found that the overexpression of triplicated genes triggered global expression dysregulation, differentially affecting transcripts, miRNAs, and proteins involved in both known and novel biological candidate pathways. Among the latter, we observed an alteration in RNA splicing, specifically modulating the expression of genes involved in cytoskeleton and axonal dynamics in DS brains. Accordingly, we found an alteration in axonal polarization in neurons from DS human iPSCs and mice. Thus, our study provides an integrated multilayer expression database capable of identifying new potential targets to aid in designing future clinical interventions for DS.
Collapse
Affiliation(s)
- Mohit Rastogi
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, Genova 16163, Italy
| | - Martina Bartolucci
- Core Facilities - Clinical Proteomics and Metabolomics, IRCCS Istituto Giannina Gaslini, Genova 16147, Italy
| | - Marina Nanni
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, Genova 16163, Italy
| | | | - Diego Vozzi
- Central RNA Laboratory, Istituto Italiano di Tecnologia, Genova 16152, Italy
| | - Andrea Petretto
- Core Facilities - Clinical Proteomics and Metabolomics, IRCCS Istituto Giannina Gaslini, Genova 16147, Italy
| | - Andrea Contestabile
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, Genova 16163, Italy.
| | - Laura Cancedda
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, Genova 16163, Italy; Dulbecco Telethon Institute, Rome 00185, Italy.
| |
Collapse
|
2
|
Murphy AJ, Wilton SD, Aung-Htut MT, McIntosh CS. Down syndrome and DYRK1A overexpression: relationships and future therapeutic directions. Front Mol Neurosci 2024; 17:1391564. [PMID: 39114642 PMCID: PMC11303307 DOI: 10.3389/fnmol.2024.1391564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 07/15/2024] [Indexed: 08/10/2024] Open
Abstract
Down syndrome is a genetic-based disorder that results from the triplication of chromosome 21, leading to an overexpression of many triplicated genes, including the gene encoding Dual-Specificity Tyrosine Phosphorylation-Regulated Kinase 1A (DYRK1A). This protein has been observed to regulate numerous cellular processes, including cell proliferation, cell functioning, differentiation, and apoptosis. Consequently, an overexpression of DYRK1A has been reported to result in cognitive impairment, a key phenotype of individuals with Down syndrome. Therefore, downregulating DYRK1A has been explored as a potential therapeutic strategy for Down syndrome, with promising results observed from in vivo mouse models and human clinical trials that administered epigallocatechin gallate. Current DYRK1A inhibitors target the protein function directly, which tends to exhibit low specificity and selectivity, making them unfeasible for clinical or research purposes. On the other hand, antisense oligonucleotides (ASOs) offer a more selective therapeutic strategy to downregulate DYRK1A expression at the gene transcript level. Advances in ASO research have led to the discovery of numerous chemical modifications that increase ASO potency, specificity, and stability. Recently, several ASOs have been approved by the U.S. Food and Drug Administration to address neuromuscular and neurological conditions, laying the foundation for future ASO therapeutics. The limitations of ASOs, including their high production cost and difficulty delivering to target tissues can be overcome by further advances in ASO design. DYRK1A targeted ASOs could be a viable therapeutic approach to improve the quality of life for individuals with Down syndrome and their families.
Collapse
Affiliation(s)
- Aidan J. Murphy
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, Australia
- Perron Institute for Neurological and Translational Science, Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Perth, WA, Australia
| | - Steve D. Wilton
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, Australia
- Perron Institute for Neurological and Translational Science, Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Perth, WA, Australia
| | - May T. Aung-Htut
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, Australia
- Perron Institute for Neurological and Translational Science, Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Perth, WA, Australia
| | - Craig S. McIntosh
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, Australia
- Perron Institute for Neurological and Translational Science, Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Perth, WA, Australia
| |
Collapse
|
3
|
Geiger M, Hurewitz SR, Pawlowski K, Baumer NT, Wilkinson CL. Alterations in aperiodic and periodic EEG activity in young children with Down syndrome. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.01.24306729. [PMID: 38746335 PMCID: PMC11092732 DOI: 10.1101/2024.05.01.24306729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Down syndrome is the most common cause of intellectual disability, yet little is known about the neurobiological pathways leading to cognitive impairments. Electroencephalographic (EEG) measures are commonly used to study neurodevelopmental disorders, but few studies have focused on young children with DS. Here we assess resting state EEG data collected from toddlers/preschoolers with DS (n=29, age 13-48 months old) and compare their aperiodic and periodic EEG features with both age-matched (n=29) and cognitive-matched (n=58) comparison groups. DS participants exhibited significantly reduced aperiodic slope, increased periodic theta power, and decreased alpha peak amplitude. A majority of DS participants displayed a prominent peak in the theta range, whereas a theta peak was not present in age-matched participants. Overall, similar findings were also observed when comparing DS and cognitive-matched groups, suggesting that EEG differences are not explained by delayed cognitive ability.
Collapse
|
4
|
Ustaoglu P, McQuarrie DWJ, Rochet A, Dix TC, Haussmann IU, Arnold R, Devaud JM, Soller M. Memory consolidation in honey bees is enhanced by down-regulation of Down syndrome cell adhesion molecule and changes its alternative splicing. Front Mol Neurosci 2024; 16:1322808. [PMID: 38264345 PMCID: PMC10803435 DOI: 10.3389/fnmol.2023.1322808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 12/13/2023] [Indexed: 01/25/2024] Open
Abstract
Down syndrome cell adhesion molecule (Dscam) gene encodes a cell adhesion molecule required for neuronal wiring. A remarkable feature of arthropod Dscam is massive alternative splicing generating thousands of different isoforms from three variable clusters of alternative exons. Dscam expression and diversity arising from alternative splicing have been studied during development, but whether they exert functions in adult brains has not been determined. Here, using honey bees, we find that Dscam expression is critically linked to memory retention as reducing expression by RNAi enhances memory after reward learning in adult worker honey bees. Moreover, alternative splicing of Dscam is altered in all three variable clusters after learning. Since identical Dscam isoforms engage in homophilic interactions, these results suggest a mechanism to alter inclusion of variable exons during memory consolidation to modify neuronal connections for memory retention.
Collapse
Affiliation(s)
- Pinar Ustaoglu
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, United Kingdom
- Birmingham Centre for Genome Biology, University of Birmingham, Birmingham, United Kingdom
| | - David W. J. McQuarrie
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, United Kingdom
- Birmingham Centre for Genome Biology, University of Birmingham, Birmingham, United Kingdom
| | - Anthony Rochet
- Research Center on Animal Cognition (CRCA), Center for Integrative Biology (CBI), CNRS, UPS, Toulouse University, Toulouse, France
| | - Thomas C. Dix
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, United Kingdom
- Birmingham Centre for Genome Biology, University of Birmingham, Birmingham, United Kingdom
| | - Irmgard U. Haussmann
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, United Kingdom
- Department of Life Science, Faculty of Health, Education and Life Sciences, Birmingham City University, Birmingham, United Kingdom
| | - Roland Arnold
- Birmingham Centre for Genome Biology, University of Birmingham, Birmingham, United Kingdom
- College of Medical and Dental Sciences, Institute of Cancer and Genomics Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Jean-Marc Devaud
- Research Center on Animal Cognition (CRCA), Center for Integrative Biology (CBI), CNRS, UPS, Toulouse University, Toulouse, France
- Institut Universitaire de France (IUF), Paris, France
| | - Matthias Soller
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, United Kingdom
- Birmingham Centre for Genome Biology, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
5
|
Perluigi M, Di Domenico F, Butterfield DA. Oxidative damage in neurodegeneration: roles in the pathogenesis and progression of Alzheimer disease. Physiol Rev 2024; 104:103-197. [PMID: 37843394 PMCID: PMC11281823 DOI: 10.1152/physrev.00030.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 03/30/2023] [Accepted: 05/24/2023] [Indexed: 10/17/2023] Open
Abstract
Alzheimer disease (AD) is associated with multiple etiologies and pathological mechanisms, among which oxidative stress (OS) appears as a major determinant. Intriguingly, OS arises in various pathways regulating brain functions, and it seems to link different hypotheses and mechanisms of AD neuropathology with high fidelity. The brain is particularly vulnerable to oxidative damage, mainly because of its unique lipid composition, resulting in an amplified cascade of redox reactions that target several cellular components/functions ultimately leading to neurodegeneration. The present review highlights the "OS hypothesis of AD," including amyloid beta-peptide-associated mechanisms, the role of lipid and protein oxidation unraveled by redox proteomics, and the antioxidant strategies that have been investigated to modulate the progression of AD. Collected studies from our groups and others have contributed to unraveling the close relationships between perturbation of redox homeostasis in the brain and AD neuropathology by elucidating redox-regulated events potentially involved in both the pathogenesis and progression of AD. However, the complexity of AD pathological mechanisms requires an in-depth understanding of several major intracellular pathways affecting redox homeostasis and relevant for brain functions. This understanding is crucial to developing pharmacological strategies targeting OS-mediated toxicity that may potentially contribute to slow AD progression as well as improve the quality of life of persons with this severe dementing disorder.
Collapse
Affiliation(s)
- Marzia Perluigi
- Department of Biochemical Sciences "A. Rossi Fanelli," Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi Fanelli," Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - D Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States
| |
Collapse
|
6
|
Koul AM, Ahmad F, Bhat A, Aein QU, Ahmad A, Reshi AA, Kaul RUR. Unraveling Down Syndrome: From Genetic Anomaly to Artificial Intelligence-Enhanced Diagnosis. Biomedicines 2023; 11:3284. [PMID: 38137507 PMCID: PMC10741860 DOI: 10.3390/biomedicines11123284] [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: 10/13/2023] [Revised: 12/04/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
Down syndrome arises from chromosomal non-disjunction during gametogenesis, resulting in an additional chromosome. This anomaly presents with intellectual impairment, growth limitations, and distinct facial features. Positive correlation exists between maternal age, particularly in advanced cases, and the global annual incidence is over 200,000 cases. Early interventions, including first and second-trimester screenings, have improved DS diagnosis and care. The manifestations of Down syndrome result from complex interactions between genetic factors linked to various health concerns. To explore recent advancements in Down syndrome research, we focus on the integration of artificial intelligence (AI) and machine learning (ML) technologies for improved diagnosis and management. Recent developments leverage AI and ML algorithms to detect subtle Down syndrome indicators across various data sources, including biological markers, facial traits, and medical images. These technologies offer potential enhancements in accuracy, particularly in cases complicated by cognitive impairments. Integration of AI and ML in Down syndrome diagnosis signifies a significant advancement in medical science. These tools hold promise for early detection, personalized treatment, and a deeper comprehension of the complex interplay between genetics and environmental factors. This review provides a comprehensive overview of neurodevelopmental and cognitive profiles, comorbidities, diagnosis, and management within the Down syndrome context. The utilization of AI and ML represents a transformative step toward enhancing early identification and tailored interventions for individuals with Down syndrome, ultimately improving their quality of life.
Collapse
Affiliation(s)
- Aabid Mustafa Koul
- Department of Immunology and Molecular Medicine, Sher-i-Kashmir Institute of Medical Sciences, Srinagar 190006, India
| | - Faisel Ahmad
- Department of Zoology, Central University of Kashmir, Ganderbal, Srinagar 190004, India
| | - Abida Bhat
- Advanced Centre for Human Genetics, Sher-i-Kashmir Institute of Medical Sciences, Srinagar 190011, India
| | - Qurat-ul Aein
- Department of Human Genetics, Guru Nanak Dev University, Amritsar 143005, Punjab, India;
| | - Ajaz Ahmad
- Departments of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Aijaz Ahmad Reshi
- Department of Computer Science, College of Computer Science and Engineering, Taibah University, Madinah 42353, Saudi Arabia;
| | - Rauf-ur-Rashid Kaul
- Department of Community Medicine, Sher-i-Kashmir Institute of Medical Sciences, Srinagar 190006, India
| |
Collapse
|
7
|
Serrano ME, Kim E, Siow B, Ma D, Rojo L, Simmons C, Hayward D, Gibbins D, Singh N, Strydom A, Fisher EM, Tybulewicz VL, Cash D. Investigating brain alterations in the Dp1Tyb mouse model of Down syndrome. Neurobiol Dis 2023; 188:106336. [PMID: 38317803 PMCID: PMC7615598 DOI: 10.1016/j.nbd.2023.106336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024] Open
Abstract
Down syndrome (DS) is one of the most common birth defects and the most prevalent genetic form of intellectual disability. DS arises from trisomy of chromosome 21, but its molecular and pathological consequences are not fully understood. In this study, we compared Dp1Tyb mice, a DS model, against their wild-type (WT) littermates of both sexes to investigate the impact of DS-related genetic abnormalities on the brain phenotype. We performed in vivo whole brain magnetic resonance imaging (MRI) and hippocampal 1H magnetic resonance spectroscopy (MRS) on the animals at 3 months of age. Subsequently, ex vivo MRI scans and histological analyses were conducted post-mortem. Our findings unveiled the following neuroanatomical and biochemical alterations in the Dp1Tyb brains: a smaller surface area and a rounder shape compared to WT brains, with DS males also presenting smaller global brain volume compared with the counterpart WT. Regional volumetric analysis revealed significant changes in 26 out of 72 examined brain regions, including the medial prefrontal cortex and dorsal hippocampus. These alterations were consistently observed in both in vivo and ex vivo imaging data. Additionally, high-resolution ex vivo imaging enabled us to investigate cerebellar layers and hippocampal sub-regions, revealing selective areas of decrease and remodelling in these structures. An analysis of hippocampal metabolites revealed an elevation in glutamine and the glutamine/glutamate ratio in the Dp1Tyb mice compared to controls, suggesting a possible imbalance in the excitation/inhibition ratio. This was accompanied by the decreased levels of taurine. Histological analysis revealed fewer neurons in the hippocampal CA3 and DG layers, along with an increase in astrocytes and microglia. These findings recapitulate multiple neuroanatomical and biochemical features associated with DS, enriching our understanding of the potential connection between chromosome 21 trisomy and the resultant phenotype.
Collapse
Affiliation(s)
- Maria Elisa Serrano
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
| | - Eugene Kim
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
| | - Bernard Siow
- The Francis Crick Institute, London, United Kingdom
| | - Da Ma
- Department of Internal Medicine Section of Gerontology and Geriatric Science, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Loreto Rojo
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
| | - Camilla Simmons
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
| | | | | | - Nisha Singh
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK
| | - Andre Strydom
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Elizabeth M.C. Fisher
- UCL Queen Square Motor Neuron Disease Centre, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, UCL, London, UK
| | | | - Diana Cash
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
| |
Collapse
|
8
|
Shaikh A, Li YQ, Lu J. Perspectives on pain in Down syndrome. Med Res Rev 2023; 43:1411-1437. [PMID: 36924439 DOI: 10.1002/med.21954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 01/08/2023] [Accepted: 02/28/2023] [Indexed: 03/18/2023]
Abstract
Down syndrome (DS) or trisomy 21 is a genetic condition often accompanied by chronic pain caused by congenital abnormalities and/or conditions, such as osteoarthritis, recurrent infections, and leukemia. Although DS patients are more susceptible to chronic pain as compared to the general population, the pain experience in these individuals may vary, attributed to the heterogenous structural and functional differences in the central nervous system, which might result in abnormal pain sensory information transduction, transmission, modulation, and perception. We tried to elaborate on some key questions and possible explanations in this review. Further clarification of the mechanisms underlying such abnormal conditions induced by the structural and functional differences is needed to help pain management in DS patients.
Collapse
Affiliation(s)
- Ammara Shaikh
- Department of Human Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning Province, China
| | - Yun-Qing Li
- Department of Anatomy, Histology, and Embryology & K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, Shaanxi Province, China
- Department of Anatomy, Basic Medical College, Zhengzhou University, Zhengzhou, China
| | - Jie Lu
- Department of Human Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
9
|
Valenti D, Vacca RA. Brain Mitochondrial Bioenergetics in Genetic Neurodevelopmental Disorders: Focus on Down, Rett and Fragile X Syndromes. Int J Mol Sci 2023; 24:12488. [PMID: 37569863 PMCID: PMC10419900 DOI: 10.3390/ijms241512488] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/31/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023] Open
Abstract
Mitochondria, far beyond their prominent role as cellular powerhouses, are complex cellular organelles active as central metabolic hubs that are capable of integrating and controlling several signaling pathways essential for neurological processes, including neurogenesis and neuroplasticity. On the other hand, mitochondria are themselves regulated from a series of signaling proteins to achieve the best efficiency in producing energy, in establishing a network and in performing their own de novo synthesis or clearance. Dysfunctions in signaling processes that control mitochondrial biogenesis, dynamics and bioenergetics are increasingly associated with impairment in brain development and involved in a wide variety of neurodevelopmental disorders. Here, we review recent evidence proving the emerging role of mitochondria as master regulators of brain bioenergetics, highlighting their control skills in brain neurodevelopment and cognition. We analyze, from a mechanistic point of view, mitochondrial bioenergetic dysfunction as causally interrelated to the origins of typical genetic intellectual disability-related neurodevelopmental disorders, such as Down, Rett and Fragile X syndromes. Finally, we discuss whether mitochondria can become therapeutic targets to improve brain development and function from a holistic perspective.
Collapse
Affiliation(s)
- Daniela Valenti
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council (CNR), Via G. Amendola 122/O, 70126 Bari, Italy
| | - Rosa Anna Vacca
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council (CNR), Via G. Amendola 122/O, 70126 Bari, Italy
| |
Collapse
|
10
|
Chen Y, Xiao Y, Zhang Y, Wang R, Wang F, Gao H, Liu Y, Zhang R, Sun H, Zhou Z, Wang S, Chen K, Sun Y, Tu M, Li J, Luo Q, Wu Y, Zhu L, Huang Y, Sun X, Guo G, Zhang D. Single-cell landscape analysis reveals systematic senescence in mammalian Down syndrome. Clin Transl Med 2023; 13:e1310. [PMID: 37461266 PMCID: PMC10352595 DOI: 10.1002/ctm2.1310] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 04/28/2023] [Accepted: 06/13/2023] [Indexed: 07/20/2023] Open
Abstract
BACKGROUND Down syndrome (DS), which is characterized by various malfunctions, is the most common chromosomal disorder. As the DS population continues to grow and most of those with DS live beyond puberty, early-onset health problems have become apparent. However, the cellular landscape and molecular alterations have not been thoroughly studied. METHODS This study utilized single-cell resolution techniques to examine DS in humans and mice, spanning seven distinct organs. A total of 71 934 mouse and 98 207 human cells were analyzed to uncover the molecular alterations occurring in different cell types and organs related to DS, specifically starting from the fetal stage. Additionally, SA-β-Gal staining, western blot, and histological study were employed to verify the alterations. RESULTS In this study, we firstly established the transcriptomic profile of the mammalian DS, deciphering the cellular map and molecular mechanism. Our analysis indicated that DS cells across various types and organs experienced senescence stresses from as early as the fetal stage. This was marked by elevated SA-β-Gal activity, overexpression of cell cycle inhibitors, augmented inflammatory responses, and a loss of cellular identity. Furthermore, we found evidence of mitochondrial disturbance, an increase in ribosomal protein transcription, and heightened apoptosis in fetal DS cells. This investigation also unearthed a regulatory network driven by an HSA21 gene, which leads to genome-wide expression changes. CONCLUSION The findings from this study offer significant insights into the molecular alterations that occur in DS, shedding light on the pathological processes underlying this disorder. These results can potentially guide future research and treatment development for DS.
Collapse
Affiliation(s)
- Yao Chen
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanyu Xiao
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanye Zhang
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Renying Wang
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Feixia Wang
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huajing Gao
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yifeng Liu
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Runju Zhang
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huiyu Sun
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Ziming Zhou
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Siwen Wang
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kai Chen
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yixi Sun
- Department of Reproductive Genetics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mixue Tu
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jingyi Li
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Birth Defect Control and Prevention Research Center of Zhejiang Province, Hangzhou, China
| | - Qiong Luo
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Birth Defect Control and Prevention Research Center of Zhejiang Province, Hangzhou, China
| | - Yiqing Wu
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Birth Defect Control and Prevention Research Center of Zhejiang Province, Hangzhou, China
| | - Linling Zhu
- Department of Gynecology, Hangzhou Women's Hospital, Hangzhou, China
| | - Yun Huang
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Birth Defect Control and Prevention Research Center of Zhejiang Province, Hangzhou, China
| | - Xiao Sun
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Guoji Guo
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Dan Zhang
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Birth Defect Control and Prevention Research Center of Zhejiang Province, Hangzhou, China
| |
Collapse
|
11
|
Kahanowitch R, Aguilar H, Weiss M, Lew J, Pan Q, Ortiz-Vergara MC, Rodriguez O, Nino G. Developmental changes in obstructive sleep apnea and sleep architecture in Down syndrome. Pediatr Pulmonol 2023; 58:1882-1888. [PMID: 37057861 DOI: 10.1002/ppul.26405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 12/19/2022] [Accepted: 03/27/2023] [Indexed: 04/15/2023]
Abstract
BACKGROUND Down syndrome (DS, also known as Trisomy 21) is a condition associated with abnormal neurodevelopment and a higher risk for sleep apnea. Our study sought to better understand and characterize the age-related developmental differences in sleep architecture and obstructive sleep apnea (OSA) severity in children with DS compared to euploid individuals. METHODS Retrospective review of polysomnograms in over 4151 infants, children, and adolescents in the pediatric sleep center at Children's National Hospital in Washington D.C. (0-18 years) including 218 individuals with DS. RESULTS The primary findings of our study are that: (1) severe OSA (obstructive apnea-hypopnea index ≥ 10/h) was more prevalent in the DS group (euploid 18% vs. DS 34%, p < 0.001) with the highest OSA severity being present in young children (<3 years old) and adolescents (>10 years old), (2) abnormalities in sleep architecture in children with DS were characterized by a prolonged rapid-eye movement (REM) sleep onset latency (SOL) (euploid 119 min vs. DS 144 min, p < 0.001) and greater arousal indexes (euploid 10.7/h vs. DS 12.2/h, p < 0.001), (3) developmental changes in the amount of REM sleep or slow wave sleep were not different in DS individuals relative to euploid children, (4) multivariate analyses showed that OSA and REM sleep latency differences between DS and euploid individuals were still present after adjusting by age, biological sex, and body mass index. CONCLUSION Severe OSA is highly prevalent in children with DS and follows an age-dependent "U" distribution with peaks in newborns/infants and children >10 years of age. Children with DS also have disturbances in sleep architecture characterized by a longer REM SOL and elevated arousal indexes. As sleep cycle generation and continuity play crucial roles in neuroplasticity and cognitive development, these findings offer clinically relevant insights to guide anticipatory guidance for infants, children, and adolescents with DS.
Collapse
Affiliation(s)
- Ryan Kahanowitch
- Division of Pediatric Pulmonary and Sleep Medicine, Children's National Hospital, George Washington University, Washington, District of Columbia, USA
| | - Hector Aguilar
- Division of Pediatric Pulmonary and Sleep Medicine, Children's National Hospital, George Washington University, Washington, District of Columbia, USA
| | - Miriam Weiss
- Division of Pediatric Pulmonary and Sleep Medicine, Children's National Hospital, George Washington University, Washington, District of Columbia, USA
| | - Jenny Lew
- Division of Pediatric Pulmonary and Sleep Medicine, Children's National Hospital, George Washington University, Washington, District of Columbia, USA
| | - Qi Pan
- Division of Pediatric Pulmonary and Sleep Medicine, Children's National Hospital, George Washington University, Washington, District of Columbia, USA
| | - Maria Camila Ortiz-Vergara
- Division of Pediatric Pulmonary and Sleep Medicine, Children's National Hospital, George Washington University, Washington, District of Columbia, USA
| | - Oscar Rodriguez
- Division of Pediatric Pulmonary and Sleep Medicine, Children's National Hospital, George Washington University, Washington, District of Columbia, USA
| | - Gustavo Nino
- Division of Pediatric Pulmonary and Sleep Medicine, Children's National Hospital, George Washington University, Washington, District of Columbia, USA
| |
Collapse
|
12
|
Watson LA, Meharena HS. From neurodevelopment to neurodegeneration: utilizing human stem cell models to gain insight into Down syndrome. Front Genet 2023; 14:1198129. [PMID: 37323671 PMCID: PMC10267712 DOI: 10.3389/fgene.2023.1198129] [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: 03/31/2023] [Accepted: 05/09/2023] [Indexed: 06/17/2023] Open
Abstract
Down syndrome (DS), caused by triplication of chromosome 21, is the most frequent aneuploidy observed in the human population and represents the most common genetic form of intellectual disability and early-onset Alzheimer's disease (AD). Individuals with DS exhibit a wide spectrum of clinical presentation, with a number of organs implicated including the neurological, immune, musculoskeletal, cardiac, and gastrointestinal systems. Decades of DS research have illuminated our understanding of the disorder, however many of the features that limit quality of life and independence of individuals with DS, including intellectual disability and early-onset dementia, remain poorly understood. This lack of knowledge of the cellular and molecular mechanisms leading to neurological features of DS has caused significant roadblocks in developing effective therapeutic strategies to improve quality of life for individuals with DS. Recent technological advances in human stem cell culture methods, genome editing approaches, and single-cell transcriptomics have provided paradigm-shifting insights into complex neurological diseases such as DS. Here, we review novel neurological disease modeling approaches, how they have been used to study DS, and what questions might be addressed in the future using these innovative tools.
Collapse
Affiliation(s)
- L. Ashley Watson
- Developmental and Cognitive Genomics Research Laboratory, Division of Biological Sciences, Section of Neurobiology, University of California, San Diego, La Jolla, CA, United States
| | - Hiruy S. Meharena
- Developmental and Cognitive Genomics Research Laboratory, Division of Biological Sciences, Section of Neurobiology, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
13
|
Liu Y, Zhang Y, Ren Z, Zeng F, Yan J. RUNX1 Upregulation Causes Mitochondrial Dysfunction via Regulating the PI3K-Akt Pathway in iPSC from Patients with Down Syndrome. Mol Cells 2023; 46:219-230. [PMID: 36625318 PMCID: PMC10086551 DOI: 10.14348/molcells.2023.2095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 09/30/2022] [Accepted: 10/03/2022] [Indexed: 01/11/2023] Open
Abstract
Down syndrome (DS) is the most common autosomal aneuploidy caused by trisomy of chromosome 21. Previous studies demonstrated that DS affected mitochondrial functions, which may be associated with the abnormal development of the nervous system in patients with DS. Runt-related transcription factor 1 (RUNX1) is an encoding gene located on chromosome 21. It has been reported that RUNX1 may affect cell apoptosis via the mitochondrial pathway. The present study investigated whether RUNX1 plays a critical role in mitochondrial dysfunction in DS and explored the mechanism by which RUNX1 affects mitochondrial functions. Expression of RUNX1 was detected in induced pluripotent stem cells of patients with DS (DS-iPSCs) and normal iPSCs (N-iPSCs), and the mitochondrial functions were investigated in the current study. Subsequently, RUNX1 was overexpressed in N-iPSCs and inhibited in DS-iPSCs. The mitochondrial functions were investigated thoroughly, including reactive oxygen species levels, mitochondrial membrane potential, ATP content and lysosomal activity. Finally, RNA-sequencing was used to explore the global expression pattern. It was observed that the expression levels of RUNX1 in DS-iPSCs were significantly higher than those in normal controls. Impaired mitochondrial functions were observed in DS-iPSCs. Of note, overexpression of RUNX1 in N-iPSCs resulted in mitochondrial dysfunction, while inhibition of RUNX1 expression could improve the mitochondrial function in DS-iPSCs. Global gene expression analysis indicated that overexpression of RUNX1 may promote the induction of apoptosis in DS-iPSCs by activating the PI3K/Akt signaling pathway. The present findings indicate that abnormal expression of RUNX1 may play a critical role in mitochondrial dysfunction in DS-iPSCs.
Collapse
Affiliation(s)
- Yanna Liu
- Shanghai Children’s Hospital, Shanghai Institute of Medical Genetics, Shanghai Jiao Tong University School of Medicine, Shanghai 200040, China
| | - Yuehua Zhang
- Shanghai Children’s Hospital, Shanghai Institute of Medical Genetics, Shanghai Jiao Tong University School of Medicine, Shanghai 200040, China
| | - Zhaorui Ren
- Shanghai Children’s Hospital, Shanghai Institute of Medical Genetics, Shanghai Jiao Tong University School of Medicine, Shanghai 200040, China
- NHC Key Laboratory of Medical Embryogenesis and Developmental Molecular Biology, Shanghai Key Laboratory of Embryo and Reproduction Engineering, Shanghai 200040, China
| | - Fanyi Zeng
- Shanghai Children’s Hospital, Shanghai Institute of Medical Genetics, Shanghai Jiao Tong University School of Medicine, Shanghai 200040, China
- NHC Key Laboratory of Medical Embryogenesis and Developmental Molecular Biology, Shanghai Key Laboratory of Embryo and Reproduction Engineering, Shanghai 200040, China
- Department of Histoembryology, Genetics & Development, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jingbin Yan
- Shanghai Children’s Hospital, Shanghai Institute of Medical Genetics, Shanghai Jiao Tong University School of Medicine, Shanghai 200040, China
- NHC Key Laboratory of Medical Embryogenesis and Developmental Molecular Biology, Shanghai Key Laboratory of Embryo and Reproduction Engineering, Shanghai 200040, China
| |
Collapse
|
14
|
Vacca RA, Augello A, Gallo L, Caggianese G, Malizia V, La Grutta S, Murero M, Valenti D, Tullo A, Balech B, Marzano F, Ghezzo A, Tancredi G, Turchetta A, Riccio MP, Bravaccio C, Scala I. Serious Games in the new era of digital-health interventions: A narrative review of their therapeutic applications to manage neurobehavior in neurodevelopmental disorders. Neurosci Biobehav Rev 2023; 149:105156. [PMID: 37019246 DOI: 10.1016/j.neubiorev.2023.105156] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/10/2023] [Accepted: 04/02/2023] [Indexed: 04/05/2023]
Abstract
Children and adolescents with neurodevelopmental disorders generally show adaptive, cognitive and motor skills impairments associated with behavioral problems, i.e., alterations in attention, anxiety and stress regulation, emotional and social relationships, which strongly limit their quality of life. This narrative review aims at providing a critical overview of the current knowledge in the field of serious games (SGs), known as digital instructional interactive videogames, applied to neurodevelopmental disorders. Indeed, a growing number of studies is drawing attention to SGs as innovative and promising interventions in managing neurobehavioral and cognitive disturbs in children with neurodevelopmental disorders. Accordingly, we provide a literature overview of the current evidence regarding the actions and the effects of SGs. In addition, we describe neurobehavioral alterations occurring in some specific neurodevelopmental disorders for which a possible therapeutic use of SGs has been suggested. Finally, we discuss findings obtained in clinical trials using SGs as digital therapeutics in neurodevelopment disorders and suggest new directions and hypotheses for future studies to bridge the gaps between clinical research and clinical practice.
Collapse
|
15
|
Abstract
Communicating the diagnosis of Down Syndrome to a couple of parents is never easy, whether before or after birth. As doctors, we must certainly rely on our own relational skills, but it is also necessary to be confident in some general indications, which are often overlooked in the strict hospital routine. This article is intended as a summary of the main articles published on this subject in the international literature, collecting and summarising the most important indications that have emerged in years of medical practice all over the world as well as in our personal experience. The diffusion of these guidelines is essential to help the doctor in this difficult task, on which there is often little training, and above all to guarantee to the parents the least traumatic communication possible.
Collapse
|
16
|
Nguyen DT, Bricout VA, Tran HT, Pham VH, Duong-Quy S. Sleep apnea in people with Down syndrome: Causes and effects of physical activity? Front Neurol 2023; 14:1123624. [PMID: 36816555 PMCID: PMC9931749 DOI: 10.3389/fneur.2023.1123624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/16/2023] [Indexed: 02/05/2023] Open
Abstract
Poor sleep quality is recognized as a major risk factor for poor health, increasing the incidence of serious chronic diseases. In people with Down syndrome, sleep apnea prevalence is significantly greater, it is caused by genetic, anatomical, endocrine, and metabolic abnormalities. The consequences of sleep disruption due to sleep apnea are very serious, especially in terms of neurocognitive and cardiovascular effects, leading to reduced life expectancy and quality of life in this population. However, the management, care, and treatment of related disorders in people with Down syndrome are still inadequate and limited. Therefore, this article wants to increase understanding and awareness about sleep apnea and the benefits of physical activity in improving sleep quality in the Down syndrome community, families, and their care specialists.
Collapse
Affiliation(s)
- Duy-Thai Nguyen
- Clinical Research Committee, Vietnam Society of Sleep Medicine (VSSM), Da Lat, Vietnam,National Institute for Control of Vaccines and Biologicals, Ministry of Health, Hanoi, Vietnam
| | | | - Hong-Tram Tran
- National Institute for Control of Vaccines and Biologicals, Ministry of Health, Hanoi, Vietnam
| | - Van-Hung Pham
- National Institute for Control of Vaccines and Biologicals, Ministry of Health, Hanoi, Vietnam
| | - Sy Duong-Quy
- Sleep Lab Centre, Bio-Medical Research Centre, Lam Dong Medical College, Da Lat, Vietnam,Immuno-Allergology Division, Hershey Medical Center, Penn State Medical College, Hershey, PA, United States,Sleep Lab Unit, Outpatient Department, Pham Ngoc Thach Medical University, Ho Chi Minh City, Vietnam,Department of Respiratory Functional Exploration, University Medical Center, University of Medicine and Pharmacy, Ho Chi Minh City, Vietnam,*Correspondence: Sy Duong-Quy ✉
| |
Collapse
|
17
|
Ganguly BB, Kadam NN. Therapeutics for mitochondrial dysfunction-linked diseases in Down syndrome. Mitochondrion 2023; 68:25-43. [PMID: 36371073 DOI: 10.1016/j.mito.2022.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 11/02/2022] [Accepted: 11/04/2022] [Indexed: 11/11/2022]
Abstract
Genome-wide deregulation contributes to mitochondrial dysfunction and impairment in oxidative phosphorylation (OXPHOS) mechanism resulting in oxidative stress, increased production of reactive oxygen species (ROS) and cell death in individuals with Down syndrome (DS). The cells, which require more energy, such as muscles, brain and heart are greatly affected. Impairment in mitochondrial network has a direct link with patho-mechanism at cellular and systemic levels at the backdrop of generalized metabolic perturbations in individuals with DS. Myriads of clinico-phenotypic features, including intellectual disability, early aging and neurodegeneration, and Alzheimer disease (AD)-related dementia are inevitable in DS-population where mitochondrial dysfunctions play the central role. Collectively, the mitochondrial abnormalities and altered energy metabolism perturbs several signaling pathways, particularly related to neurogenesis, which are directly associated with cognitive development and early onset of AD in individuals with DS. Therefore, therapeutic challenges for amelioration of the mitochondrial defects were perceived to improve the quality of life of the DS population. A number of pharmacologically active natural compounds such as polyphenols, antioxidants and flavonoids have shown convincing outcome for reversal of the dysfunctional mitochondrial network and oxidative metabolism, and improvement in intellectual skill in mouse models of DS and humans with DS.
Collapse
Affiliation(s)
- Bani Bandana Ganguly
- MGM New Bombay Hospital and MGM Institute of Health Sciences, Navi Mumbai, India.
| | - Nitin N Kadam
- MGM New Bombay Hospital and MGM Institute of Health Sciences, Navi Mumbai, India
| |
Collapse
|
18
|
Behl T, Rana T, Sehgal A, Makeen HA, Albratty M, Alhazmi HA, Meraya AM, Bhatia S, Sachdeva M. Phytochemicals targeting nitric oxide signaling in neurodegenerative diseases. Nitric Oxide 2023; 130:1-11. [PMID: 36375788 DOI: 10.1016/j.niox.2022.11.001] [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: 09/27/2022] [Revised: 10/25/2022] [Accepted: 11/07/2022] [Indexed: 11/13/2022]
Abstract
Neurodegenerative diseases are a set of diseases in which slow and progressive neuronal loss occurs. Nitric oxide (NO) as a neurotransmitter performs key roles in the stimulation and blockade of various inflammatory processes. Although physiological NO is necessary for protection against a variety of pathogens, reactive oxygen species-mediated oxidative stress induces inflammatory cascades and apoptosis. Activation of glial cells particularly astrocytes and microglia induce overproduction of NO, resulting in neuroinflammation and neurodegenerative disorders. Hence, inhibiting the overproduction of NO is a beneficial therapeutic approach for numerous neuroinflammatory conditions. Several compounds have been explored for the management of neurodegenerative disorders, but they have minor symptomatic benefits and several adverse effects. Phytochemicals have currently gained more consideration owing to their ability to reduce the overproduction of NO in neurodegenerative disorders. Furthermore, phytochemicals are generally considered to be safe and beneficial. The mechanisms of NO generation and their implications in neurodegenerative disorders are explored in this review article, as well as several newly discovered phytochemicals that might have NO inhibitory activity. The current review could aid in the discovery of new anti-neuroinflammatory drugs that can suppress NO generation, particularly during neuroinflammatory and neurodegenerative conditions.
Collapse
Affiliation(s)
- Tapan Behl
- School of Health Sciences and Technology, University of Petroleum and Energy Studies, Bidholi, Dehradun, India.
| | - Tarapati Rana
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Aayush Sehgal
- GHG Khalsa College of Pharmacy, Gurusar Sadhar, Punjab, India
| | - Hafiz A Makeen
- Pharmacy Practice Research Unit, Clinical Pharmacy Department, College of Pharmacy, Jazan University, Saudi Arabia
| | - Mohammed Albratty
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Hassan A Alhazmi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan, Saudi Arabia; Substance Abuse and Toxicology Research Center, Jazan University, Jazan, Saudi Arabia
| | - Abdulkarim M Meraya
- Pharmacy Practice Research Unit, Department of Clinical Pharmacy, College of Pharmacy, Jazan University, Saudi Arabia
| | - Saurabh Bhatia
- Natural & Medical Sciences Research Centre, University of Nizwa, Birkat Al Mauz, Nizwa, Oman
| | - Monika Sachdeva
- Fatima College of Health Science, Al Ain, United Arab Emirates
| |
Collapse
|
19
|
Singla RK, De R, Efferth T, Mezzetti B, Sahab Uddin M, Ntie-Kang F, Wang D, Schultz F, Kharat KR, Devkota HP, Battino M, Sur D, Lordan R, Patnaik SS, Tsagkaris C, Sai CS, Tripathi SK, Găman MA, Ahmed MEO, González-Burgos E, Babiaka SB, Paswan SK, Odimegwu JI, Akram F, Simal-Gandara J, Urquiza MS, Tikhonov A, Mondal H, Singla S, Lonardo SD, Mulholland EJ, Cenanovic M, Maigoro AY, Giampieri F, Lee S, Tzvetkov NT, Louka AM, Verma P, Chopra H, Olea SP, Khan J, Alvarez Suarez JM, Zheng X, Tomczyk M, Sabnani MK, Medina CDV, Khalid GM, Boyina HK, Georgiev MI, Supuran CT, Sobarzo-Sánchez E, Fan TP, Pittala V, Sureda A, Braidy N, Russo GL, Vacca RA, Banach M, Lizard G, Zarrouk A, Hammami S, Orhan IE, Aggarwal BB, Perry G, Miller MJ, Heinrich M, Bishayee A, Kijjoa A, Arkells N, Bredt D, Wink M, Fiebich BL, Kiran G, Yeung AWK, Gupta GK, Santini A, Lucarini M, Durazzo A, El-Demerdash A, Dinkova-Kostova AT, Cifuentes A, Souto EB, Zubair MAM, Badhe P, Echeverría J, Horbańczuk JO, Horbanczuk OK, Sheridan H, Sheshe SM, Witkowska AM, Abu-Reidah IM, Riaz M, Ullah H, Oladipupo AR, Lopez V, Sethiya NK, Shrestha BG, Ravanan P, Gupta SC, Alzahrani QE, Dama Sreedhar P, Xiao J, Moosavi MA, Subramani PA, Singh AK, Chettupalli AK, Patra JK, Singh G, Karpiński TM, Al-Rimawi F, Abiri R, Ahmed AF, Barreca D, Vats S, Amrani S, Fimognari C, Mocan A, Hritcu L, Semwal P, Shiblur Rahaman M, Emerald M, Akinrinde AS, Singh A, Joshi A, Joshi T, Khan SY, Balla GOA, Lu A, Pai SR, Ghzaiel I, Acar N, Es-Safi NE, Zengin G, Kureshi AA, Sharma AK, Baral B, Rani N, Jeandet P, Gulati M, Kapoor B, Mohanta YK, Emam-Djomeh Z, Onuku R, Depew JR, Atrooz OM, Goh BH, Andrade JC, Konwar B, Shine VJ, Ferreira JMLD, Ahmad J, Chaturvedi VK, Skalicka-Woźniak K, Sharma R, Gautam RK, Granica S, Parisi S, Kumar R, Atanasov AG, Shen B. The International Natural Product Sciences Taskforce (INPST) and the power of Twitter networking exemplified through #INPST hashtag analysis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 108:154520. [PMID: 36334386 DOI: 10.1016/j.phymed.2022.154520] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 07/12/2022] [Accepted: 10/21/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND The development of digital technologies and the evolution of open innovation approaches have enabled the creation of diverse virtual organizations and enterprises coordinating their activities primarily online. The open innovation platform titled "International Natural Product Sciences Taskforce" (INPST) was established in 2018, to bring together in collaborative environment individuals and organizations interested in natural product scientific research, and to empower their interactions by using digital communication tools. METHODS In this work, we present a general overview of INPST activities and showcase the specific use of Twitter as a powerful networking tool that was used to host a one-week "2021 INPST Twitter Networking Event" (spanning from 31st May 2021 to 6th June 2021) based on the application of the Twitter hashtag #INPST. RESULTS AND CONCLUSION The use of this hashtag during the networking event period was analyzed with Symplur Signals (https://www.symplur.com/), revealing a total of 6,036 tweets, shared by 686 users, which generated a total of 65,004,773 impressions (views of the respective tweets). This networking event's achieved high visibility and participation rate showcases a convincing example of how this social media platform can be used as a highly effective tool to host virtual Twitter-based international biomedical research events.
Collapse
Affiliation(s)
- Rajeev K Singla
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Xinchuan Road 2222, Chengdu, Sichuan, China; School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab-144411, India
| | - Ronita De
- ICMR-National Institute of Cholera and Enteric Diseases, P-33, CIT Rd, Subhas Sarobar Park, Phool Bagan, Beleghata, Kolkata, West Bengal 700010, India
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany
| | - Bruno Mezzetti
- Department of Agriculture, Food and Environmental Sciences (D3A) Università Politecnica Delle Marche Ancona, IT, Italy
| | - Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh; Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Fidele Ntie-Kang
- Department of Chemistry, Faculty of Science, University of Buea, Buea P.O. Box 63, Cameroon
| | - Dongdong Wang
- Centre for Metabolism, Obesity, and Diabetes Research, Department of Medicine, McMaster University, HSC 4N71, 1280 Main Street West, Hamilton, ON L8S 4K1, Canada
| | - Fabien Schultz
- Technical University of Berlin, Institute of Biotechnology, Faculty III - Process Sciences, Gustav-Meyer-Allee 25, Berlin 13355, Germany; Neubrandenburg University of Applied Sciences, Department of Agriculture and Food Sciences, Brodaer Str. 2, Neubrandenburg 17033, Germany
| | | | - Hari Prasad Devkota
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1Oe-honmachi, Kumamoto 862-0973, Japan; Program for Leading Graduate Schools, HIGO Program, Kumamoto University, Japan
| | - Maurizio Battino
- Department of Clinical Sciences, Faculty of Medicine, Polytechnic University of Marche, Ancona 60131, Italy; International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China
| | - Daniel Sur
- Department of Medical Oncology, "Iuliu Hatieganu" University of Medicine and Pharmacy Cluj-Napoca, Romania
| | - Ronan Lordan
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, PA, United States
| | - Sourav S Patnaik
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, United States
| | | | - Chandragiri Siva Sai
- Amity Institute of Pharmacy, Amity University, Uttar Pradesh, Lucknow Campus, Gomati Nagar, Lucknow, Uttar Pradesh 226010, India
| | - Surya Kant Tripathi
- Cancer Drug Resistance Laboratory, National Institute of Technology Rourkela, Odisha-769008, India
| | - Mihnea-Alexandru Găman
- ″Carol Davila" University of Medicine and Pharmacy, 8 Eroii Sanitari Boulevard, Bucharest, Romania; Center of Hematology and Bone Marrow Transplantation, Fundeni Clinical Institute, 258 Fundeni Road, Bucharest, Romania
| | - Mosa E O Ahmed
- Department of Pharmacognosy, Faculty of Pharmacy, Al Neelain University, Khartoum, Sudan
| | - Elena González-Burgos
- Department of Pharmacology, Pharmacognosy and Botany, University Complutense of Madrid, Spain
| | - Smith B Babiaka
- Department of Chemistry, Faculty of Science, University of Buea, Buea P.O. Box 63, Cameroon
| | | | | | - Faizan Akram
- Bahawalpur College of Pharmacy (BCP), Bahawalpur Medical and Dental College (BMDC), Bahawalpur, Pakistan
| | - Jesus Simal-Gandara
- Universidade de Vigo, Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Faculty of Science, Ourense E-32004, Spain
| | | | - Aleksei Tikhonov
- Translational Research Laboratory in Immunotherapy, Gustave Roussy, Villejuif, France
| | - Himel Mondal
- Department of Physiology, All India Institute of Medical Sciences, Deoghar, Jharkhand, India
| | - Shailja Singla
- iGlobal Research and Publishing Foundation, New Delhi, India
| | - Sara Di Lonardo
- Research Institute on Terrestrial Ecosystems-Italian National Research Council (IRET-CNR), Via Madonna del Piano 10, Sesto Fiorentino Fi 50019, Italy
| | - Eoghan J Mulholland
- Gastrointestinal Stem Cell Biology Laboratory, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom; Somerville College, University of Oxford, Oxford, United Kingdom
| | | | | | - Francesca Giampieri
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia; Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Santander, Spain
| | - Soojin Lee
- Department of Bioscience and Biotechnology, Chungnam National University, Republic of Korea
| | - Nikolay T Tzvetkov
- Department of Biochemical Pharmacology and Drug Design, Institute of Molecular Biology "Roumen Tsanev", Bulgarian Academy of Sciences, Bulgaria
| | | | - Pritt Verma
- Department of Pharmacology, CSIR-NBRI, Lucknow, India
| | - Hitesh Chopra
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | | | - Johra Khan
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah 11952, Saudi Arabia
| | - José M Alvarez Suarez
- Departamento de Ingeniería en Alimentos, Colegio de Ciencias e Ingenierías, Universidad San Francisco de Quito, Quito, Ecuador
| | - Xiaonan Zheng
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Michał Tomczyk
- Department of Pharmacognosy, Faculty of Pharmacy with the Division of Laboratory Medicine, Medical University of Białystok, ul. Mickiewicza 2a, Białystok 15-230, Poland
| | - Manoj Kumar Sabnani
- The University of Texas at Arlington, United States; Alloy Therapeutics, United States
| | | | - Garba M Khalid
- Pharmaceutical Engineering Group, School of Pharmacy, Queen's University, Belfast BT9, United Kingdom
| | - Hemanth Kumar Boyina
- School of Pharmacy, Department of Pharmacology, Anurag University, Venkatapur, Medchal, Hyderabad, Telangana 500088, India
| | - Milen I Georgiev
- Laboratory of Metabolomics, Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, 139 Ruski Blvd., Plovdiv 4000, Bulgaria
| | | | - Eduardo Sobarzo-Sánchez
- Instituto de Investigación y Postgrado, Facultad de Ciencias de la Salud, Universidad Central de Chile, Santiago 8330507, Chile; Department of Organic Chemistry, Faculty of Pharmacy, University of Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Tai-Ping Fan
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Science and Medicine, Northwest University, Xi'an, China
| | - Valeria Pittala
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
| | - Antoni Sureda
- Research Group in Community Nutrition and Oxidative Stress, University of the Balearic Islands-IUNICS, Health Research Institute of Balearic Islands (IdISBa), and CIBEROBN (Physiopathology of Obesity and Nutrition), Palma, Balearic Islands E-07122, Spain
| | - Nady Braidy
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, University of New South Wales, Sydney, Australia
| | - Gian Luigi Russo
- National Research Council, Institute of Food Sciences, Avellino 83100, Italy
| | - Rosa Anna Vacca
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Council of Research, Bari 70126, Italy
| | - Maciej Banach
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz (MUL), Lodz, Poland; Cardiovascular Research Centre, University of Zielona Gora, Zielona Gora, Poland
| | - Gérard Lizard
- Université de Bourgogne / Inserm, Laboratoire Bio-PeroxIL, Faculté des Sciences Gabriel, 6 Boulevard Gabriel, Dijon 21000 France
| | - Amira Zarrouk
- University of Monastir (Tunisia), Faculty of Medicine, LR-NAFS 'Nutrition - Functional Food & Vascular Health', Tunisia
| | - Sonia Hammami
- University of Monastir (Tunisia), Faculty of Medicine, LR-NAFS 'Nutrition - Functional Food & Vascular Health', Tunisia
| | - Ilkay Erdogan Orhan
- Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, Ankara 06330, Türkiye
| | | | - George Perry
- Department of Neuroscience, Developmental, and Regenerative Biology, University of Texas, United States
| | | | | | - Anupam Bishayee
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, United States
| | - Anake Kijjoa
- Instituto de Ciências Biomédicas Abel Salazar e CIIMAR, Universidade do Porto, Portugal
| | - Nicolas Arkells
- International Natural Product Sciences Taskforce (INSPT), United States
| | | | - Michael Wink
- Heidelberg University, Institute of Pharmacy and Molecular Biotechnology (IPMB), Heidelberg 69120, Germany
| | - Bernd L Fiebich
- Neurochemistry and Neuroimmunology Research Group, Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | - Andy Wai Kan Yeung
- Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, University of Hong Kong, Hong Kong, China
| | - Girish Kumar Gupta
- Department of Pharmaceutical Chemistry, Sri Sai College of Pharmacy, Badhani, Pathankot, Punjab, India
| | - Antonello Santini
- University of Napoli Federico II, Department of Pharmacy. Via D Montesano 49, Napoli 80131, Italy
| | - Massimo Lucarini
- CREA-Research Centre for Food and Nutrition, Via Ardeatina 546 00178 Rome, Italy
| | - Alessandra Durazzo
- CREA-Research Centre for Food and Nutrition, Via Ardeatina 546 00178 Rome, Italy
| | - Amr El-Demerdash
- Metabolic Biology & Biological Chemistry Department, John Innes Centre, Norwich Research Park, Norwich NR4 7UH, United Kingdom; Organic Chemistry Division, Chemistry Department, Faculty of Science, Mansoura University, Mansoura 35516, Egypt
| | | | | | - Eliana B Souto
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, Porto 4050-313, Portugal
| | | | - Pravin Badhe
- Swalife Foundation, India; Swalife Biotech Ltd, Ireland; Sinhgad College of Pharmacy, Vadgaon (BK) Pune Maharashtra India
| | - Javier Echeverría
- Departamento de Ciencias del Ambiente, Facultad de Química y Biología, Universidad de Santiago de Chile, Chile
| | - Jarosław Olav Horbańczuk
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzębiec 05-552, Poland
| | - Olaf K Horbanczuk
- Department of Technique and Food Product Development, Warsaw University of Life Sciences (WULS-SGGW) 159c Nowoursynowska, Warsaw 02-776, Poland
| | - Helen Sheridan
- The NatPro Centre. Trinity College Dublin. Dublin 2, Ireland
| | | | | | - Ibrahim M Abu-Reidah
- School of Science and the Environment, Grenfell Campus, Memorial University of Newfoundland, Corner Brook A2H 5G4, Canada
| | - Muhammad Riaz
- Department of Pharmacy, Shaheed Benazir Bhutto University, Sheringal 18050, Pakistan
| | - Hammad Ullah
- Department of Pharmacy, University of Naples Federico II, Naples 80131, Italy
| | - Akolade R Oladipupo
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Lagos, Nigeria; Department of Chemistry, Nelson Mandela University, Port Elizabeth, South Africa
| | - Víctor Lopez
- Department of Pharmacy, Universidad San Jorge, Villanueva de Gállego (Zaragoza), Spain
| | | | | | - Palaniyandi Ravanan
- Department of Microbiology, School of Life Sciences, Central University of Tamil Nadu, Thiruvarur, India
| | - Subash Chandra Gupta
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, India; Department of Biochemistry, All India Institute of Medical Sciences, Guwahati, Assam, India
| | - Qushmua E Alzahrani
- Department of Pharmacy/Nursing Medicine Health and Environment, University of the Region of Joinville (UNIVILLE) Brazil, Sana Catarina, Joinville, Brazil
| | | | | | - Mohammad Amin Moosavi
- Molecular Medicine Department, Institute of Medical Biotechnology, National Institute of Genetics Engineering and Biotechnology, Tehran P.O. Box: 14965/161, Iran
| | - Parasuraman Aiya Subramani
- Independent Researcher, Vels Institute of Science, Technology and Advanced Studies (VISTAS), Chennai, India - 600048. formerly, Pallavaram, Chennai 600117, India
| | - Amit Kumar Singh
- Department of Biochemistry, University of Allahabad, Prayagraj 211002 India
| | | | - Jayanta Kumar Patra
- Research Institute of Integrative Life Sciences, Dongguk University-Seoul, Goyangsi 10326, Republic of Korea
| | - Gopal Singh
- Department of Plant Functional Metabolomics, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | - Tomasz M Karpiński
- Chair and Department of Medical Microbiology, Poznań University of Medical Sciences, Wieniawskiego 3, Poznań 61-712, Poland
| | | | - Rambod Abiri
- Department of Forestry Science and Biodiversity, Faculty of Forestry and Environment, Universiti Putra Malaysia, UPM Serdang, Selangor 43400, Malaysia
| | - Atallah F Ahmed
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Davide Barreca
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, Università degli Studi di Messina, Messina, Italy
| | - Sharad Vats
- Department of Bioscience and Biotechnology, Banasthali Vidyapith, Rajasthan 304022, India
| | - Said Amrani
- Laboratoire de Biologie et de Physiologie des Organismes, Faculté des Sciences Biologiques, USTHB, Bab Ezzouar, Alger, Algeria
| | | | - Andrei Mocan
- Department of Pharmaceutical Botany, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Lucian Hritcu
- Department of Biology, Alexandru Ioan Cuza University of Iasi, Bd. Carol I, No. 11, Iasi 700506, Romania
| | - Prabhakar Semwal
- Department of Life Sciences, Graphic Era Deemed to be University, Dehradun, Uttarakhand 248002, India
| | - Md Shiblur Rahaman
- Department of Environmental and Preventive Medicine, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Mila Emerald
- PHYTOCEUTICALS International™ & NOVOTEK Global Solutions™, Canada
| | - Akinleye Stephen Akinrinde
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | | | - Ashima Joshi
- Sardar Bhagwan Singh University, Balawala, Dehradun, India
| | - Tanuj Joshi
- Department of Pharmaceutical Sciences, Bhimtal, Kumaun University (Nainital), India
| | - Shafaat Yar Khan
- Research Lab III, Hematology & Vascular Biology, Department of Zoology, University of Sargodha, Sargodha, Pakistan
| | - Gareeballah Osman Adam Balla
- Department of Pharmacology, College of Veterinary Medicine, Sudan University of Science and Technology, Hilat Kuku, Khartoum North P.O. Box No. 204, Sudan
| | - Aiping Lu
- School of Chinese Medicine, Hong Kong Baptist University, HongKong, China
| | - Sandeep Ramchandra Pai
- Department of Botany, Rayat Shikshan Sanstha's, Dada Patil Mahavidyalaya, Karjat, Maharashtra, India
| | - Imen Ghzaiel
- Université de Bourgogne, Inserm, Laboratoire Bio - PeroxIL, Faculté des Sciences Gabriel, 6 Boulevard Gabriel, Dijon 21000 France; University Tunis El Manar, Tunis, Tunisia
| | | | - Nour Eddine Es-Safi
- Mohammed V University in Rabat, LPCMIO, Materials Science Center (MSC), Ecole Normale Supérieure, Rabat, Morocco
| | - Gokhan Zengin
- Department of Biology, Science Faculty, Selcuk University, Konya, Turkey
| | - Azazahemad A Kureshi
- Department of Chemistry, Sardar Vallabhbhai National Institute of Technology, Surat, India
| | | | | | - Neeraj Rani
- Department of Pharmaceutical Sciences, Chaudhary Bansilal University, Bhiwani, Haryana, India
| | - Philippe Jeandet
- University of Reims, Research Unit Induced Resistance and Plant Bioprotection, USC INRAe 1488, Reims, France
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T. Road (NH 1) Phagwara, Punjab 144411 India
| | - Bhupinder Kapoor
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T. Road (NH 1) Phagwara, Punjab 144411 India
| | - Yugal Kishore Mohanta
- Department of Applied Biology, School of Biological Sciences, University of Science and Technology Meghalaya (USTM), Techno City, Kling Road, Baridua, Ri-Bhoi, Meghalaya 793101, India
| | | | - Raphael Onuku
- Department of Pharmaceutical and Medicinal Chemistry, Faculty of Pharmaceutical Sciences, University of Nigeria, Nigeria
| | | | - Omar M Atrooz
- Department of Biological Sciences, Mutah University, Jordan
| | - Bey Hing Goh
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China; Biofunctional Molecule Exploratory (BMEX) Research Group, School of Pharmacy, Monash University Malaysia, Subang Jaya, Malaysia
| | - Jose Carlos Andrade
- TOXRUN - Toxicology Research Unit, University Institute of Health Sciences, CESPU, Gandra, Portugal
| | | | - V J Shine
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, Kerala 695014, India
| | | | - Jamil Ahmad
- Department of Human Nutrition, The University of Agriculture Peshawar, Khyber Pakhtunkhwa, Pakistan
| | - Vivek K Chaturvedi
- Department of Gastroenterology, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | | | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Rupesh K Gautam
- Deparment of Pharmacology, Indore Institute of Pharmacy, IIST Campus, Rau-Indore-453331, India
| | - Sebastian Granica
- Microbiota Lab, Department of Pharmacognosy and Molecular Basis of Phytotherapy, Medical University of Warsaw, Poland
| | - Salvatore Parisi
- Lourdes Matha Institute of Hotel Management and Catering Technology, Kerala State, India
| | - Rishabh Kumar
- School of Medical and Allied Sciences, K.R. Mangalam University, Sohna Road, Gurugram, Haryana 122103, India
| | - Atanas G Atanasov
- Ludwig Boltzmann Institute for Digital Health and Patient Safety, Medical University of Vienna, Spitalgasse 23, Vienna 1090, Austria; Department of Pharmaceutical Sciences, University of Vienna, Althanstraße 14, Vienna 1090, Austria; Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzebiec, Magdalenka 05-552, Poland.
| | - Bairong Shen
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Xinchuan Road 2222, Chengdu, Sichuan, China.
| |
Collapse
|
20
|
Choudhary N, Tewari D, Nabavi SF, Kashani HRK, Lorigooini Z, Filosa R, Khan FB, Masoudian N, Nabavi SM. Plant based food bioactives: A boon or bane for neurological disorders. Crit Rev Food Sci Nutr 2022; 64:3279-3325. [PMID: 36369694 DOI: 10.1080/10408398.2022.2131729] [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: 11/14/2022]
Abstract
Neurological disorders are the foremost occurring diseases across the globe resulting in progressive dysfunction, loss of neuronal structure ultimately cell death. Therefore, attention has been drawn toward the natural resources for the search of neuroprotective agents. Plant-based food bioactives have emerged as potential neuroprotective agents for the treatment of neurodegenerative disorders. This comprehensive review primarily focuses on various plant food bioactive, mechanisms, therapeutic targets, in vitro and in vivo studies in the treatment of neurological disorders to explore whether they are boon or bane for neurological disorders. In addition, the clinical perspective of plant food bioactives in neurological disorders are also highlighted. Scientific evidences point toward the enormous therapeutic efficacy of plant food bioactives in the prevention or treatment of neurological disorders. Nevertheless, identification of food bioactive components accountable for the neuroprotective effects, mechanism, clinical trials, and consolidation of information flow are warranted. Plant food bioactives primarily act by mediating through various pathways including oxidative stress, neuroinflammation, apoptosis, excitotoxicity, specific proteins, mitochondrial dysfunction, and reversing neurodegeneration and can be used for the prevention and therapy of neurodegenerative disorders. In conclusion, the plant based food bioactives are boon for neurological disorders.
Collapse
Affiliation(s)
- Neeraj Choudhary
- Department of Pharmacognosy, Adesh Institute of Pharmacy and Biomedical Sciences, Adesh University, Bathinda, Punjab, India
| | - Devesh Tewari
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, India
| | - Seyed Fazel Nabavi
- Advanced Medical Pharma (AMP-Biotec), Biopharmaceutical Innovation Centre Via Cortenocera, 82030, San Salvatore Telesino, (BN), Italy
- Nutringredientes Research Center, Federal Institute of Education, Science and Technology (IFCE), Baturite, Ceara, Brazil
| | - Hamid Reza Khayat Kashani
- Department of Neurosurgery, Imam Hossein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Lorigooini
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Rosanna Filosa
- Advanced Medical Pharma (AMP-Biotec), Biopharmaceutical Innovation Centre Via Cortenocera, 82030, San Salvatore Telesino, (BN), Italy
- Department of Science and Technology, University of Sannio, 82100, Benevento, Italy
| | - Farheen Badrealam Khan
- Department of Biology, College of Science, The United Arab Emirates University, Al Ain, 15551 United Arab Emirates
| | - Nooshin Masoudian
- Advanced Medical Pharma (AMP-Biotec), Biopharmaceutical Innovation Centre Via Cortenocera, 82030, San Salvatore Telesino, (BN), Italy
| | - Seyed Mohammad Nabavi
- Advanced Medical Pharma (AMP-Biotec), Biopharmaceutical Innovation Centre Via Cortenocera, 82030, San Salvatore Telesino, (BN), Italy
- Nutringredientes Research Center, Federal Institute of Education, Science and Technology (IFCE), Baturite, Ceara, Brazil
| |
Collapse
|
21
|
von Scheibler EN, van Eeghen AM, de Koning TJ, Kuijf ML, Zinkstok JR, Müller AR, van Amelsvoort TA, Boot E. Parkinsonism in Genetic Neurodevelopmental Disorders: A Systematic Review. Mov Disord Clin Pract 2022; 10:17-31. [PMID: 36699000 PMCID: PMC9847320 DOI: 10.1002/mdc3.13577] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 06/25/2022] [Accepted: 08/16/2022] [Indexed: 01/28/2023] Open
Abstract
Background With advances in clinical genetic testing, associations between genetic neurodevelopmental disorders and parkinsonism are increasingly recognized. In this review, we aimed to provide a comprehensive overview of reports on parkinsonism in genetic neurodevelopmental disorders and summarize findings related to genetic diagnosis, clinical features and proposed disease mechanisms. Methods A systematic literature review was conducted in PubMed and Embase on June 15, 2021. Search terms for parkinsonism and genetic neurodevelopmental disorders, using generic terms and the Human Phenotype Ontology, were combined. Study characteristics and descriptive data were extracted from the articles using a modified version of the Cochrane Consumers and Communication Review Group's data extraction template. The protocol was registered in PROSPERO (CRD42020191035). Results The literature search yielded 208 reports for data-extraction, describing 69 genetic disorders in 422 patients. The five most reported from most to least frequent were: 22q11.2 deletion syndrome, beta-propeller protein-associated neurodegeneration, Down syndrome, cerebrotendinous xanthomatosis, and Rett syndrome. Notable findings were an almost equal male to female ratio, an early median age of motor onset (26 years old) and rigidity being more common than rest tremor. Results of dopaminergic imaging and response to antiparkinsonian medication often supported the neurodegenerative nature of parkinsonism. Moreover, neuropathology results showed neuronal loss in the majority of cases. Proposed disease mechanisms included aberrant mitochondrial function and disruptions in neurotransmitter metabolism, endosomal trafficking, and the autophagic-lysosomal and ubiquitin-proteasome system. Conclusion Parkinsonism has been reported in many GNDs. Findings from this study may provide clues for further research and improve management of patients with GNDs and/or parkinsonism.
Collapse
Affiliation(s)
- Emma N.M.M. von Scheibler
- Advisium'sHeeren Loo ZorggroepAmersfoortThe Netherlands,Department of Psychiatry and NeuropsychologyMaastricht UniversityMaastrichtThe Netherlands
| | - Agnies M. van Eeghen
- Advisium'sHeeren Loo ZorggroepAmersfoortThe Netherlands,Emma Children's HospitalUniversity of AmsterdamAmsterdamThe Netherlands
| | - Tom J. de Koning
- Department of GeneticsUniversity of GroningenGroningenThe Netherlands,Expertise Centre Movement Disorders GroningenUniversity Medical Centre GroningenGroningenThe Netherlands,Pediatrics, Department of Clinical SciencesLund UniversityLundSweden
| | - Mark L. Kuijf
- Department of NeurologyMaastricht University Medical CentreMaastrichtThe Netherlands
| | - Janneke R. Zinkstok
- Department of PsychiatryRadoud University Medical CentreNijmegenThe Netherlands,Karakter child and adolescent psychiatryNijmegenThe Netherlands,Department of Psychiatry and Brain CenterUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Annelieke R. Müller
- Advisium'sHeeren Loo ZorggroepAmersfoortThe Netherlands,Emma Children's HospitalUniversity of AmsterdamAmsterdamThe Netherlands
| | | | - Erik Boot
- Advisium'sHeeren Loo ZorggroepAmersfoortThe Netherlands,Department of Psychiatry and NeuropsychologyMaastricht UniversityMaastrichtThe Netherlands,The Dalglish Family 22q ClinicUniversity Health NetworkTorontoOntarioCanada
| |
Collapse
|
22
|
Fatty Acids: A Safe Tool for Improving Neurodevelopmental Alterations in Down Syndrome? Nutrients 2022; 14:nu14142880. [PMID: 35889838 PMCID: PMC9323400 DOI: 10.3390/nu14142880] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 02/06/2023] Open
Abstract
The triplication of chromosome 21 causes Down syndrome (DS), a genetic disorder that is characterized by intellectual disability (ID). The causes of ID start in utero, leading to impairments in neurogenesis, and continue into infancy, leading to impairments in dendritogenesis, spinogenesis, and connectivity. These defects are associated with alterations in mitochondrial and metabolic functions and precocious aging, leading to the early development of Alzheimer’s disease. Intense efforts are currently underway, taking advantage of DS mouse models to discover pharmacotherapies for the neurodevelopmental and cognitive deficits of DS. Many treatments that proved effective in mouse models may raise safety concerns over human use, especially at early life stages. Accumulating evidence shows that fatty acids, which are nutrients present in normal diets, exert numerous positive effects on the brain. Here, we review (i) the knowledge obtained from animal models regarding the effects of fatty acids on the brain, by focusing on alterations that are particularly prominent in DS, and (ii) the progress recently made in a DS mouse model, suggesting that fatty acids may indeed represent a useful treatment for DS. This scenario should prompt the scientific community to further explore the potential benefit of fatty acids for people with DS.
Collapse
|
23
|
Kalhori MR, Soleimani M, Yari K, Moradi M, Kalhori AA. MiR-1290: a potential therapeutic target for regenerative medicine or diagnosis and treatment of non-malignant diseases. Clin Exp Med 2022:10.1007/s10238-022-00854-9. [PMID: 35802264 DOI: 10.1007/s10238-022-00854-9] [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: 04/19/2022] [Accepted: 06/20/2022] [Indexed: 11/03/2022]
Abstract
MicroRNAs are a set of small non-coding RNAs that could change gene expression with post-transcriptional regulation. MiRNAs have a significant role in regulating molecular signaling pathways and innate and adaptive immune system activity. Moreover, miRNAs can be utilized as a powerful instrument for tissue engineers and regenerative medicine by altering the expression of genes and growth factors. MiR-1290, which was first discovered in human embryonic stem cells, is one of those miRNAs that play an essential role in developing the fetal nervous system. This review aims to discuss current findings on miR-1290 in different human pathologies and determine whether manipulation of miR-1290 could be considered a possible therapeutic strategy to treat different non-malignant diseases. The results of these studies suggest that the regulation of miR-1290 may be helpful in the treatment of some bacterial (leprosy) and viral infections (HIV, influenza A, and Borna disease virus). Also, adjusting the expression of miR-1290 in non-infectious diseases such as celiac disease, necrotizing enterocolitis, polycystic ovary syndrome, pulmonary fibrosis, ankylosing spondylitis, muscle atrophy, sarcopenia, and ischemic heart disease can help to treat these diseases better. In addition to acting as a biomarker for the diagnosis of non-malignant diseases (such as NAFLD, fetal growth, preeclampsia, down syndrome, chronic rhinosinusitis, and oral lichen planus), the miR-1290 can also be used as a valuable instrument in tissue engineering and reconstructive medicine. Consequently, it is suggested that the regulation of miR-1290 could be considered a possible therapeutic target in the treatment of non-malignant diseases in the future.
Collapse
Affiliation(s)
- Mohammad Reza Kalhori
- Regenerative Medicine Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Masoud Soleimani
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Kheirollah Yari
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mahmoudreza Moradi
- Regenerative Medicine Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Amir Ali Kalhori
- Regenerative Medicine Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
24
|
Stagni F, Bartesaghi R. The Challenging Pathway of Treatment for Neurogenesis Impairment in Down Syndrome: Achievements and Perspectives. Front Cell Neurosci 2022; 16:903729. [PMID: 35634470 PMCID: PMC9130961 DOI: 10.3389/fncel.2022.903729] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 04/19/2022] [Indexed: 12/17/2022] Open
Abstract
Down syndrome (DS), also known as trisomy 21, is a genetic disorder caused by triplication of Chromosome 21. Gene triplication may compromise different body functions but invariably impairs intellectual abilities starting from infancy. Moreover, after the fourth decade of life people with DS are likely to develop Alzheimer’s disease. Neurogenesis impairment during fetal life stages and dendritic pathology emerging in early infancy are thought to be key determinants of alterations in brain functioning in DS. Although the progressive improvement in medical care has led to a notable increase in life expectancy for people with DS, there are currently no treatments for intellectual disability. Increasing evidence in mouse models of DS reveals that pharmacological interventions in the embryonic and neonatal periods may greatly benefit brain development and cognitive performance. The most striking results have been obtained with pharmacotherapies during embryonic life stages, indicating that it is possible to pharmacologically rescue the severe neurodevelopmental defects linked to the trisomic condition. These findings provide hope that similar benefits may be possible for people with DS. This review summarizes current knowledge regarding (i) the scope and timeline of neurogenesis (and dendritic) alterations in DS, in order to delineate suitable windows for treatment; (ii) the role of triplicated genes that are most likely to be the key determinants of these alterations, in order to highlight possible therapeutic targets; and (iii) prenatal and neonatal treatments that have proved to be effective in mouse models, in order to rationalize the choice of treatment for human application. Based on this body of evidence we will discuss prospects and challenges for fetal therapy in individuals with DS as a potential means of drastically counteracting the deleterious effects of gene triplication.
Collapse
Affiliation(s)
- Fiorenza Stagni
- Department for Life Quality Studies, University of Bologna, Rimini, Italy
| | - Renata Bartesaghi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
- *Correspondence: Renata Bartesaghi,
| |
Collapse
|
25
|
Biselli JM, Zampieri BL, Biselli-Chicote PM, de Souza JES, Bürger MC, da Silva WA, Goloni-Bertollo EM, Pavarino ÉC. Differential microRNA expression profile in blood of children with Down syndrome suggests a role in immunological dysfunction. Hum Cell 2022; 35:639-648. [PMID: 35060072 PMCID: PMC8773395 DOI: 10.1007/s13577-022-00672-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 01/09/2022] [Indexed: 11/18/2022]
Abstract
Down syndrome (DS), caused by trisomy of chromosome 21 (HSA21), results in a broad range of phenotypes. However, the determinants contributing to the complex and variable phenotypic expression of DS are still not fully known. Changes in microRNAs (miRNAs), short non-coding RNA molecules that regulate gene expression post-transcriptionally, have been associated with some DS phenotypes. Here, we investigated the genome-wide mature miRNA expression profile in peripheral blood mononuclear cells (PBMCs) of children with DS and controls and identified biological processes and pathways relevant to the DS pathogenesis. The expression of 754 mature miRNAs was profiled in PBMCs from six children with DS and six controls by RT-qPCR using TaqMan® Array Human MicroRNA Cards. Functions and signaling pathways analyses were performed using DIANA-miRPath v.3 and DIANA-microT-CDS software. Children with DS presented six differentially expressed miRNAs (DEmiRs): four overexpressed (miR-378a-3p, miR-130b-5p, miR-942-5p, and miR-424-3p) and two downregulated (miR-452-5p and miR-668-3p). HSA21-derived miRNAs investigated were not found to be differentially expressed between the groups. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses showed potential target genes involved in biological processes and pathways pertinent to immune response, e.g., toll-like receptors (TLRs) signaling, Hippo, and transforming growth factor β (TGF-β) signaling pathways. These results suggest that altered miRNA expression could be contributing to the well-known immunological dysfunction observed in individuals with DS.
Collapse
Affiliation(s)
- Joice Matos Biselli
- Department of Molecular Biology, Faculdade de Medicina de São José Do Rio Preto , Genetics and Molecular Biology Research Unit (UPGEM), São José Do Rio Preto Medical School (FAMERP), Avenida Brigadeiro Faria Lima, nº 5416 - UPGEM/Bloco U-6, CEP: 15.090-000, São José Do Rio Preto, São Paulo, Brazil
| | - Bruna Lancia Zampieri
- Department of Molecular Biology, Faculdade de Medicina de São José Do Rio Preto , Genetics and Molecular Biology Research Unit (UPGEM), São José Do Rio Preto Medical School (FAMERP), Avenida Brigadeiro Faria Lima, nº 5416 - UPGEM/Bloco U-6, CEP: 15.090-000, São José Do Rio Preto, São Paulo, Brazil
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Patrícia Matos Biselli-Chicote
- Department of Molecular Biology, Faculdade de Medicina de São José Do Rio Preto , Genetics and Molecular Biology Research Unit (UPGEM), São José Do Rio Preto Medical School (FAMERP), Avenida Brigadeiro Faria Lima, nº 5416 - UPGEM/Bloco U-6, CEP: 15.090-000, São José Do Rio Preto, São Paulo, Brazil
| | - Jorge Estefano Santana de Souza
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
- Metrópole Digital Institute (IMD), UFRN, Natal, RN, Brazil
- National Institute of Science and Technology in Stem Cell and Cell Therapy and Center for Cell Based Therapy, Ribeirao Preto, São Paulo, Brazil
| | - Matheus Carvalho Bürger
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
- National Institute of Science and Technology in Stem Cell and Cell Therapy and Center for Cell Based Therapy, Ribeirao Preto, São Paulo, Brazil
| | - Wilson Araújo da Silva
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
- National Institute of Science and Technology in Stem Cell and Cell Therapy and Center for Cell Based Therapy, Ribeirao Preto, São Paulo, Brazil
| | - Eny Maria Goloni-Bertollo
- Department of Molecular Biology, Faculdade de Medicina de São José Do Rio Preto , Genetics and Molecular Biology Research Unit (UPGEM), São José Do Rio Preto Medical School (FAMERP), Avenida Brigadeiro Faria Lima, nº 5416 - UPGEM/Bloco U-6, CEP: 15.090-000, São José Do Rio Preto, São Paulo, Brazil
| | - Érika Cristina Pavarino
- Department of Molecular Biology, Faculdade de Medicina de São José Do Rio Preto , Genetics and Molecular Biology Research Unit (UPGEM), São José Do Rio Preto Medical School (FAMERP), Avenida Brigadeiro Faria Lima, nº 5416 - UPGEM/Bloco U-6, CEP: 15.090-000, São José Do Rio Preto, São Paulo, Brazil.
| |
Collapse
|
26
|
Bartesaghi R, Vicari S, Mobley WC. Prenatal and Postnatal Pharmacotherapy in Down Syndrome: The Search to Prevent or Ameliorate Neurodevelopmental and Neurodegenerative Disorders. Annu Rev Pharmacol Toxicol 2022; 62:211-233. [PMID: 34990205 PMCID: PMC9632639 DOI: 10.1146/annurev-pharmtox-041521-103641] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Those with Down syndrome (DS)-trisomy for chromosome 21-are routinely impacted by cognitive dysfunction and behavioral challenges in children and adults and Alzheimer's disease in older adults. No proven treatments specifically address these cognitive or behavioral changes. However, advances in the establishment of rodent models and human cell models promise to support development of such treatments. A research agenda that emphasizes the identification of overexpressed genes that contribute demonstrably to abnormalities in cognition and behavior in model systems constitutes a rational next step. Normalizing expression of such genes may usher in an era of successful treatments applicable across the life span for those with DS.
Collapse
Affiliation(s)
- Renata Bartesaghi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Stefano Vicari
- Department of Life Sciences and Public Health, Catholic University of the Sacred Heart, 00168 Rome, Italy,Child and Adolescent Neuropsychiatry Unit, Department of Neuroscience, Bambino Gesù Children’s Hospital, IRCCS, 00165-00146 Rome, Italy
| | - William C. Mobley
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093, USA
| |
Collapse
|
27
|
Schworer EK, Esbensen AJ, Fidler DJ, Beebe DW, Carle A, Wiley S. Evaluating working memory outcome measures for children with Down syndrome. JOURNAL OF INTELLECTUAL DISABILITY RESEARCH : JIDR 2022; 66:195-211. [PMID: 33763953 PMCID: PMC8463631 DOI: 10.1111/jir.12833] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 01/13/2021] [Accepted: 03/02/2021] [Indexed: 05/13/2023]
Abstract
BACKGROUND There is a critical need for the psychometric evaluation of outcome measures to be used in clinical trials targeting cognition in Down syndrome (DS). This study examines a specific cognitive skill that is of particular importance in DS, working memory, and the psychometric properties of a set of standardised measurements to assess working memory in individuals with DS. METHODS Ninety children and adolescents ages 6 to 18 years old with DS were assessed on a selection of verbal and visuospatial working memory subtests of standardised clinical assessments at two time points to examine feasibility, distributional qualities, test-retest reliability and convergent validity against a priori criteria. Caregivers also completed an adaptive behaviour questionnaire to address working memory subtests' associations with broader developmental functioning. RESULTS The Stanford Binet-5 Verbal Working Memory, Differential Ability Scales-2 Recognition of Pictures, Stanford Binet-5 Nonverbal Working Memory and Wechsler Intelligence Scale for Children-5 Picture Span measures met the most psychometric criteria overall across the full age and IQ range of the study. Although Differential Ability Scales-2 Recall of Sequential Order and Differential Ability Scales-2 Recall of Digits Backward met the fewest a priori criteria, follow-up analyses suggested greater feasibility in specific age and IQ ranges. CONCLUSIONS Several working memory measures appear to be psychometrically sound and appropriate for use in clinical trials for children with DS, especially when focusing on raw scores. However, floor effects on standard scores and feasibility of some measures were problematic. Guidelines for use of the working memory subtests with this population are provided.
Collapse
Affiliation(s)
- Emily K. Schworer
- Division of Developmental and Behavioral Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Anna J. Esbensen
- Division of Developmental and Behavioral Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Deborah J. Fidler
- Human Development and Family Studies, Colorado State University, Fort Collins, CO, USA
| | - Dean W. Beebe
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Behavioral Medicine and Clinical Psychology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Adam Carle
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- James M Anderson Center for Health Systems Excellence, Cincinnati, OH, USA
- Department of Psychology, University of Cincinnati, Cincinnati, OH, USA
| | - Susan Wiley
- Division of Developmental and Behavioral Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
28
|
Valenti D, Stagni F, Emili M, Guidi S, Bartesaghi R, Vacca RA. Impaired Brain Mitochondrial Bioenergetics in the Ts65Dn Mouse Model of Down Syndrome Is Restored by Neonatal Treatment with the Polyphenol 7,8-Dihydroxyflavone. Antioxidants (Basel) 2021; 11:antiox11010062. [PMID: 35052567 PMCID: PMC8773005 DOI: 10.3390/antiox11010062] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/22/2021] [Accepted: 12/24/2021] [Indexed: 12/18/2022] Open
Abstract
Down syndrome (DS), a major genetic cause of intellectual disability, is characterized by numerous neurodevelopmental defects. Previous in vitro studies highlighted a relationship between bioenergetic dysfunction and reduced neurogenesis in progenitor cells from the Ts65Dn mouse model of DS, suggesting a critical role of mitochondrial dysfunction in neurodevelopmental alterations in DS. Recent in vivo studies in Ts65Dn mice showed that neonatal supplementation (Days P3–P15) with the polyphenol 7,8-dihydroxyflavone (7,8-DHF) fully restored hippocampal neurogenesis. The current study was aimed to establish whether brain mitochondrial bioenergetic defects are already present in Ts65Dn pups and whether early treatment with 7,8-DHF positively impacts on mitochondrial function. In the brain and cerebellum of P3 and P15 Ts65Dn pups we found a strong impairment in the oxidative phosphorylation apparatus, resulting in a deficit in mitochondrial ATP production and ATP content. Administration of 7,8-DHF (dose: 5 mg/kg/day) during Days P3–P15 fully restored bioenergetic dysfunction in Ts65Dn mice, reduced the levels of oxygen radicals and reinstated the hippocampal levels of PGC-1α. No pharmacotherapy is available for DS. From current findings, 7,8-DHF emerges as a treatment with a good translational potential for improving mitochondrial bioenergetics and, thus, mitochondria-linked neurodevelopmental alterations in DS.
Collapse
Affiliation(s)
- Daniela Valenti
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council (CNR), 70126 Bari, Italy;
- Correspondence: (D.V.); (R.B.)
| | - Fiorenza Stagni
- Department for Life Quality Studies, University of Bologna, 47921 Rimini, Italy;
| | - Marco Emili
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy; (M.E.); (S.G.)
| | - Sandra Guidi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy; (M.E.); (S.G.)
| | - Renata Bartesaghi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy; (M.E.); (S.G.)
- Correspondence: (D.V.); (R.B.)
| | - Rosa Anna Vacca
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council (CNR), 70126 Bari, Italy;
| |
Collapse
|
29
|
Structure of cell-cell adhesion mediated by the Down syndrome cell adhesion molecule. Proc Natl Acad Sci U S A 2021; 118:2022442118. [PMID: 34531300 DOI: 10.1073/pnas.2022442118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/09/2021] [Indexed: 11/18/2022] Open
Abstract
The Down syndrome cell adhesion molecule (DSCAM) belongs to the immunoglobulin superfamily (IgSF) and plays important roles in neural development. It has a large ectodomain, including 10 Ig-like domains and 6 fibronectin III (FnIII) domains. Previous data have shown that DSCAM can mediate cell adhesion by forming homophilic dimers between cells and contributes to self-avoidance of neurites or neuronal tiling, which is important for neural network formation. However, the organization and assembly of DSCAM at cell adhesion interfaces has not been fully understood. Here we combine electron microscopy and other biophysical methods to characterize the structure of the DSCAM-mediated cell adhesion and generate three-dimensional views of the adhesion interfaces of DSCAM by electron tomography. The results show that mouse DSCAM forms a regular pattern at the adhesion interfaces. The Ig-like domains contribute to both trans homophilic interactions and cis assembly of the pattern, and the FnIII domains are crucial for the cis pattern formation as well as the interaction with the cell membrane. By contrast, no obvious assembly pattern is observed at the adhesion interfaces mediated by mouse DSCAML1 or Drosophila DSCAMs, suggesting the different structural roles and mechanisms of DSCAMs in mediating cell adhesion and neural network formation.
Collapse
|
30
|
Ahmed MM, Johnson NR, Boyd TD, Coughlan C, Chial HJ, Potter H. Innate Immune System Activation and Neuroinflammation in Down Syndrome and Neurodegeneration: Therapeutic Targets or Partners? Front Aging Neurosci 2021; 13:718426. [PMID: 34603007 PMCID: PMC8481947 DOI: 10.3389/fnagi.2021.718426] [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] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 08/16/2021] [Indexed: 12/24/2022] Open
Abstract
Innate immune system activation and inflammation are associated with and may contribute to clinical outcomes in people with Down syndrome (DS), neurodegenerative diseases such as Alzheimer's disease (AD), and normal aging. In addition to serving as potential diagnostic biomarkers, innate immune system activation and inflammation may play a contributing or causal role in these conditions, leading to the hypothesis that effective therapies should seek to dampen their effects. However, recent intervention studies with the innate immune system activator granulocyte-macrophage colony-stimulating factor (GM-CSF) in animal models of DS, AD, and normal aging, and in an AD clinical trial suggest that activating the innate immune system and inflammation may instead be therapeutic. We consider evidence that DS, AD, and normal aging are accompanied by innate immune system activation and inflammation and discuss whether and when during the disease process it may be therapeutically beneficial to suppress or promote such activation.
Collapse
Affiliation(s)
- Md. Mahiuddin Ahmed
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- University of Colorado Alzheimer’s and Cognition Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Noah R. Johnson
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- University of Colorado Alzheimer’s and Cognition Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Timothy D. Boyd
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- University of Colorado Alzheimer’s and Cognition Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Partner Therapeutics, Inc., Lexington, MA, United States
| | - Christina Coughlan
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- University of Colorado Alzheimer’s and Cognition Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Heidi J. Chial
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- University of Colorado Alzheimer’s and Cognition Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Huntington Potter
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- University of Colorado Alzheimer’s and Cognition Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
31
|
Amin RE, Abdel-Meguid IE, El-Refaie NM, Fakher W, El-Tabie D, El-Gindy HA. Clinical Profiles, Congenital Heart Disease, and Other Comorbidities Among Egyptian Children with Down Syndrome: A Tertiary Center Study. JOURNAL OF CHILD SCIENCE 2021. [DOI: 10.1055/s-0041-1735537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Abstract
Introduction Down syndrome (DS) is the most common chromosomal disorder. It is accompanied by several comorbidities, which could lead to severe morbidity and mortality. Congenital heart disease (CHD) is one of the most commonly described condition.
Objective This study aimed to determine clinical profiles, dysmorphic features, CHD, and DS associated comorbidities in a tertiary center (Cairo, Egypt).
Patients and Methods This descriptive study included 290 patients diagnosed with DS, who presented to the Clinical Genetics clinic, Cairo University Children Hospitals, from February 2018 to December 2019. The patients' ages ranged from 2 to 4 years old. All patients were evaluated by full history, clinical examination, anthropometric measurements, and assessment of developmental milestones. Patients' diagnostic investigations including karyotype, thyroid function, and echocardiography were checked.
Results The study population consisted of 290 children with DS of which 196 (67.6%) were male, 115 (40%) had CHD, the most prevalent atrial septal defect (ASD), patent ductus arteriosus (PDA), and ventricular septal defect (VSD) accounting for 10.7, 7.1, and 4.2%, respectively. Common dysmorphic features were upward slanting palpebral fissures (98.6%), hypertelorism (97.9%), and sandal gap (60.7%). Thyroid dysfunction was the second prevalent comorbidity, found in 35 patients (12.1%). Global developmental delay was reported affecting language (99%), motor (94.8%), and social (92.8%) domains.
Conclusion The prevalence of CHD among children with DS was 40% with ASD, PDA, and VSD being the commonest. Thyroid dysfunction was the second most common comorbidity. The most prevalent dysmorphic features were upward slanting palpebral fissures, hypertelorism, and sandal gap. Developmental delay was very common, language being the most affected domain.
Collapse
Affiliation(s)
- Radwa Ezzat Amin
- Department of Pediatrics, Faculty of Medicine, Cairo University, Cairo, Egypt
| | | | | | - Walaa Fakher
- Department of Psychiatry, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Dina El-Tabie
- Department of Psychiatry, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Hala Ahmed El-Gindy
- Department of Pediatrics, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
32
|
Toshikawa H, Ikenaka A, Li L, Nishinaka-Arai Y, Niwa A, Ashida A, Kazuki Y, Nakahata T, Tamai H, Russell DW, Saito MK. N-Acetylcysteine prevents amyloid-β secretion in neurons derived from human pluripotent stem cells with trisomy 21. Sci Rep 2021; 11:17377. [PMID: 34462463 PMCID: PMC8405674 DOI: 10.1038/s41598-021-96697-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 08/10/2021] [Indexed: 11/09/2022] Open
Abstract
Down syndrome (DS) is caused by the trisomy of chromosome 21. Among the many disabilities found in individuals with DS is an increased risk of early-onset Alzheimer's disease (AD). Although higher oxidative stress and an upregulation of amyloid β (Aβ) peptides from an extra copy of the APP gene are attributed to the AD susceptibility, the relationship between the two factors is unclear. To address this issue, we established an in vitro cellular model using neurons differentiated from DS patient-derived induced pluripotent stem cells (iPSCs) and isogenic euploid iPSCs. Neurons differentiated from DS patient-derived iPSCs secreted more Aβ compared to those differentiated from the euploid iPSCs. Treatment of the neurons with an antioxidant, N-acetylcysteine, significantly suppressed the Aβ secretion. These findings suggest that oxidative stress has an important role in controlling the Aβ level in neurons differentiated from DS patient-derived iPSCs and that N-acetylcysteine can be a potential therapeutic option to ameliorate the Aβ secretion.
Collapse
Affiliation(s)
- Hiromitsu Toshikawa
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan.,Osaka Medical and Pharmaceutical University, Takatsuki, 5690801, Japan.,Social Welfare Organization "SAISEIKAI" Imperial Gift Foundation Inc., Saiseikai Suita Hospital, Suita, 5640013, Japan
| | - Akihiro Ikenaka
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Li Li
- Division of Hematology, School of Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Yoko Nishinaka-Arai
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan.,Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, 6068507, Japan
| | - Akira Niwa
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Akira Ashida
- Osaka Medical and Pharmaceutical University, Takatsuki, 5690801, Japan
| | - Yasuhiro Kazuki
- Chromosome Engineering Research Center, Tottori University, Tottori, Japan.,Division of Genome and Cellular Functions, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine, Tottori University, Tottori, Japan
| | - Tatsutoshi Nakahata
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Hiroshi Tamai
- Osaka Medical and Pharmaceutical University, Takatsuki, 5690801, Japan.,Institute for Developmental Brain Research, Osaka Medical and Pharmaceutical University, Takatsuki, 5690801, Japan
| | - David W Russell
- Division of Hematology, School of Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Megumu K Saito
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan.
| |
Collapse
|
33
|
The Alteration of Chloride Homeostasis/GABAergic Signaling in Brain Disorders: Could Oxidative Stress Play a Role? Antioxidants (Basel) 2021; 10:antiox10081316. [PMID: 34439564 PMCID: PMC8389245 DOI: 10.3390/antiox10081316] [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] [Received: 08/02/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 12/22/2022] Open
Abstract
In neuronal precursors and immature neurons, the depolarizing (excitatory) effect of γ-Aminobutyric acid (GABA) signaling is associated with elevated [Cl−]i; as brain cells mature, a developmental switch occurs, leading to the decrease of [Cl−]i and to the hyperpolarizing (inhibitory) effect of GABAergic signaling. [Cl−]i is controlled by two chloride co-transporters: NKCC1, which causes Cl− to accumulate into the cells, and KCC2, which extrudes it. The ontogenetic upregulation of the latter determines the above-outlined switch; however, many other factors contribute to the correct [Cl−]i in mature neurons. The dysregulation of chloride homeostasis is involved in seizure generation and has been associated with schizophrenia, Down’s Syndrome, Autism Spectrum Disorder, and other neurodevelopmental disorders. Recently, much effort has been put into developing new drugs intended to inhibit NKCC1 activity, while no attention has been paid to the origin of [Cl−]i dysregulation. Our study examines the pathophysiology of Cl− homeostasis and focuses on the impact of oxidative stress (OS) and inflammation on the activity of Cl− co-transporters, highlighting the relevance of OS in numerous brain abnormalities and diseases. This hypothesis supports the importance of primary prevention during pregnancy. It also integrates the therapeutic framework addressed to restore normal GABAergic signaling by counteracting the alteration in chloride homeostasis in central nervous system (CNS) cells, aiming at limiting the use of drugs that potentially pose a health risk.
Collapse
|
34
|
Valenti D, Vacca RA, Moro L, Atlante A. Mitochondria Can Cross Cell Boundaries: An Overview of the Biological Relevance, Pathophysiological Implications and Therapeutic Perspectives of Intercellular Mitochondrial Transfer. Int J Mol Sci 2021; 22:8312. [PMID: 34361078 PMCID: PMC8347886 DOI: 10.3390/ijms22158312] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/19/2021] [Accepted: 07/28/2021] [Indexed: 01/07/2023] Open
Abstract
Mitochondria are complex intracellular organelles traditionally identified as the powerhouses of eukaryotic cells due to their central role in bioenergetic metabolism. In recent decades, the growing interest in mitochondria research has revealed that these multifunctional organelles are more than just the cell powerhouses, playing many other key roles as signaling platforms that regulate cell metabolism, proliferation, death and immunological response. As key regulators, mitochondria, when dysfunctional, are involved in the pathogenesis of a wide range of metabolic, neurodegenerative, immune and neoplastic disorders. Far more recently, mitochondria attracted renewed attention from the scientific community for their ability of intercellular translocation that can involve whole mitochondria, mitochondrial genome or other mitochondrial components. The intercellular transport of mitochondria, defined as horizontal mitochondrial transfer, can occur in mammalian cells both in vitro and in vivo, and in physiological and pathological conditions. Mitochondrial transfer can provide an exogenous mitochondrial source, replenishing dysfunctional mitochondria, thereby improving mitochondrial faults or, as in in the case of tumor cells, changing their functional skills and response to chemotherapy. In this review, we will provide an overview of the state of the art of the up-to-date knowledge on intercellular trafficking of mitochondria by discussing its biological relevance, mode and mechanisms underlying the process and its involvement in different pathophysiological contexts, highlighting its therapeutic potential for diseases with mitochondrial dysfunction primarily involved in their pathogenesis.
Collapse
Affiliation(s)
- Daniela Valenti
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM)-CNR, Via G. Amendola122/O, 70126 Bari, Italy; (R.A.V.); (L.M.)
| | | | | | - Anna Atlante
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM)-CNR, Via G. Amendola122/O, 70126 Bari, Italy; (R.A.V.); (L.M.)
| |
Collapse
|
35
|
D'Antoni S, de Bari L, Valenti D, Borro M, Bonaccorso CM, Simmaco M, Vacca RA, Catania MV. Aberrant mitochondrial bioenergetics in the cerebral cortex of the Fmr1 knockout mouse model of fragile X syndrome. Biol Chem 2021; 401:497-503. [PMID: 31702995 DOI: 10.1515/hsz-2019-0221] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 10/14/2019] [Indexed: 12/12/2022]
Abstract
Impaired energy metabolism may play a role in the pathogenesis of neurodevelopmental disorders including fragile X syndrome (FXS). We checked brain energy status and some aspects of cell bioenergetics, namely the activity of key glycolytic enzymes, glycerol-3-phosphate shuttle and mitochondrial respiratory chain (MRC) complexes, in the cerebral cortex of the Fmr1 knockout (KO) mouse model of FXS. We found that, despite a hyperactivation of MRC complexes, adenosine triphosphate (ATP) production via mitochondrial oxidative phosphorylation (OXPHOS) is compromised, resulting in brain energy impairment in juvenile and late-adult Fmr1 KO mice. Thus, an altered mitochondrial energy metabolism may contribute to neurological impairment in FXS.
Collapse
Affiliation(s)
- Simona D'Antoni
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), Via Paolo Gaifami 18, I-95126 Catania, Italy
| | - Lidia de Bari
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), CNR, Via Giovanni Amendola 165/A, I-70126 Bari, Italy
| | - Daniela Valenti
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), CNR, Via Giovanni Amendola 165/A, I-70126 Bari, Italy
| | - Marina Borro
- Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Sapienza University of Rome, Via di Grottarossa 1035, I-00189 Rome, Italy
| | | | - Maurizio Simmaco
- Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Sapienza University of Rome, Via di Grottarossa 1035, I-00189 Rome, Italy
| | - Rosa Anna Vacca
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), CNR, Via Giovanni Amendola 165/A, I-70126 Bari, Italy
| | - Maria Vincenza Catania
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), Via Paolo Gaifami 18, I-95126 Catania, Italy.,Oasi Research Institute - IRCCS, Via Conte Ruggero 73, I-94018 Troina, Italy
| |
Collapse
|
36
|
Kawatani K, Nambara T, Nawa N, Yoshimatsu H, Kusakabe H, Hirata K, Tanave A, Sumiyama K, Banno K, Taniguchi H, Arahori H, Ozono K, Kitabatake Y. A human isogenic iPSC-derived cell line panel identifies major regulators of aberrant astrocyte proliferation in Down syndrome. Commun Biol 2021; 4:730. [PMID: 34127780 PMCID: PMC8203796 DOI: 10.1038/s42003-021-02242-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 05/18/2021] [Indexed: 12/15/2022] Open
Abstract
Astrocytes exert adverse effects on the brains of individuals with Down syndrome (DS). Although a neurogenic-to-gliogenic shift in the fate-specification step has been reported, the mechanisms and key regulators underlying the accelerated proliferation of astrocyte precursor cells (APCs) in DS remain elusive. Here, we established a human isogenic cell line panel based on DS-specific induced pluripotent stem cells, the XIST-mediated transcriptional silencing system in trisomic chromosome 21, and genome/chromosome-editing technologies to eliminate phenotypic fluctuations caused by genetic variation. The transcriptional responses of genes observed upon XIST induction and/or downregulation are not uniform, and only a small subset of genes show a characteristic expression pattern, which is consistent with the proliferative phenotypes of DS APCs. Comparative analysis and experimental verification using gene modification reveal dose-dependent proliferation-promoting activity of DYRK1A and PIGP on DS APCs. Our collection of human isogenic cell lines provides a comprehensive set of cellular models for further DS investigations. Keiji Kawatani et al. developed a panel of Down syndrome (DS) isogenic astrocytes derived from iPSCs to observe the consequence of DS on astrocyte precursor proliferation, differentiation, and gene expression. Their results suggest a dose-dependent effect of DYRK1A and PIGP on DS-derived astrocyte precursor proliferation, and represent a valuable resource and cellular model for future DS research.
Collapse
Affiliation(s)
- Keiji Kawatani
- Department of Pediatrics, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Toshihiko Nambara
- Department of Pediatrics, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Nobutoshi Nawa
- Department of Pediatrics, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Hidetaka Yoshimatsu
- Department of Pediatrics, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Haruna Kusakabe
- Department of Pediatrics, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Katsuya Hirata
- Department of Pediatrics, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.,Department of Neonatal Medicine, Osaka Women's and Children's Hospital, Izumi, Osaka, Japan
| | - Akira Tanave
- Laboratory for Mouse Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
| | - Kenta Sumiyama
- Laboratory for Mouse Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
| | - Kimihiko Banno
- Department of Pediatrics, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.,Department of Physiology II, Nara Medical University, Kashihara, Nara, Japan
| | - Hidetoshi Taniguchi
- Department of Pediatrics, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Hitomi Arahori
- Department of Pediatrics, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Keiichi Ozono
- Department of Pediatrics, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Yasuji Kitabatake
- Department of Pediatrics, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.
| |
Collapse
|
37
|
Emmerzaal TL, Nijkamp G, Veldic M, Rahman S, Andreazza AC, Morava E, Rodenburg RJ, Kozicz T. Effect of neuropsychiatric medications on mitochondrial function: For better or for worse. Neurosci Biobehav Rev 2021; 127:555-571. [PMID: 34000348 DOI: 10.1016/j.neubiorev.2021.05.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/12/2021] [Accepted: 05/04/2021] [Indexed: 01/22/2023]
Abstract
Individuals with mitochondrial disease often present with psychopathological comorbidity, and mitochondrial dysfunction has been proposed as the underlying pathobiology in various psychiatric disorders. Several studies have suggested that medications used to treat neuropsychiatric disorders could directly influence mitochondrial function. This review provides a comprehensive overview of the effect of these medications on mitochondrial function. We collected preclinical information on six major groups of antidepressants and other neuropsychiatric medications and found that the majority of these medications either positively influenced mitochondrial function or showed mixed effects. Only amitriptyline, escitalopram, and haloperidol were identified as having exclusively adverse effects on mitochondrial function. In the absence of formal clinical trials, and until such trials are completed, the data from preclinical studies reported and discussed here could inform medication prescribing practices for individuals with psychopathology and impaired mitochondrial function in the underlying pathology.
Collapse
Affiliation(s)
- Tim L Emmerzaal
- Radboud University Medical Center, Donders Institute for Brain Cognition and Behaviour, Department of Medical Imaging, Anatomy, Nijmegen, The Netherlands; Mayo Clinic, Department of Clinical Genomics, Rochester, MN, USA
| | - Gerben Nijkamp
- Radboud University Medical Center, Donders Institute for Brain Cognition and Behaviour, Department of Medical Imaging, Anatomy, Nijmegen, The Netherlands
| | - Marin Veldic
- Mayo Clinic, Department of Psychiatry, Rochester, MN, USA
| | - Shamima Rahman
- Mitochondrial Research Group, UCL Great Ormond Street Institute of Child Health, London, United Kingdom; Metabolic Unit, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Ana Cristina Andreazza
- University of Toronto, Temerty Faculty of Medicine, Department of Pharmacology & Toxicology and Psychiatry, Toronto, Canada
| | - Eva Morava
- Mayo Clinic, Department of Clinical Genomics, Rochester, MN, USA; Mayo Clinic, Department of Laboratory Medicine and Pathology, Rochester, MN, USA
| | - Richard J Rodenburg
- Radboud Center for Mitochondrial Medicine, Translational Metabolic Laboratory, Department of Pediatrics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Tamas Kozicz
- Radboud University Medical Center, Donders Institute for Brain Cognition and Behaviour, Department of Medical Imaging, Anatomy, Nijmegen, The Netherlands; Mayo Clinic, Department of Clinical Genomics, Rochester, MN, USA; Mayo Clinic, Department of Biochemistry and Molecular Biology, Rochester, MN, USA.
| |
Collapse
|
38
|
Bayona-Bafaluy MP, Garrido-Pérez N, Meade P, Iglesias E, Jiménez-Salvador I, Montoya J, Martínez-Cué C, Ruiz-Pesini E. Down syndrome is an oxidative phosphorylation disorder. Redox Biol 2021; 41:101871. [PMID: 33540295 PMCID: PMC7859316 DOI: 10.1016/j.redox.2021.101871] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/29/2020] [Accepted: 01/13/2021] [Indexed: 02/07/2023] Open
Abstract
Down syndrome is the most common genomic disorder of intellectual disability and is caused by trisomy of chromosome 21. Several genes in this chromosome repress mitochondrial biogenesis. The goal of this study was to evaluate whether early overexpression of these genes may cause a prenatal impairment of oxidative phosphorylation negatively affecting neurogenesis. Reduction in the mitochondrial energy production and a lower mitochondrial function have been reported in diverse tissues or cell types, and also at any age, including early fetuses, suggesting that a defect in oxidative phosphorylation is an early and general event in Down syndrome individuals. Moreover, many of the medical conditions associated with Down syndrome are also frequently found in patients with oxidative phosphorylation disease. Several drugs that enhance mitochondrial biogenesis are nowadays available and some of them have been already tested in mouse models of Down syndrome restoring neurogenesis and cognitive defects. Because neurogenesis relies on a correct mitochondrial function and critical periods of brain development occur mainly in the prenatal and early neonatal stages, therapeutic approaches intended to improve oxidative phosphorylation should be provided in these periods.
Collapse
Affiliation(s)
- M Pilar Bayona-Bafaluy
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, C/ Miguel Servet, 177. 50013, Zaragoza, Spain and C/ Pedro Cerbuna, 12, 50009, Zaragoza, Spain; Instituto de Investigación Sanitaria (IIS) de Aragón, Av. San Juan Bosco, 13, 50009, Zaragoza, Spain; Centro de Investigaciones Biomédicas en Rd de Enfermedades Raras (CIBERER), Av. Monforte de Lemos, 3-5, 28029, Madrid, Spain; Instituto de Biocomputación y Física de Sistemas Complejos (BIFI), Universidad de Zaragoza. C/ Mariano Esquillor (Edificio I+D), 50018, Zaragoza, Spain.
| | - Nuria Garrido-Pérez
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, C/ Miguel Servet, 177. 50013, Zaragoza, Spain and C/ Pedro Cerbuna, 12, 50009, Zaragoza, Spain; Instituto de Investigación Sanitaria (IIS) de Aragón, Av. San Juan Bosco, 13, 50009, Zaragoza, Spain; Centro de Investigaciones Biomédicas en Rd de Enfermedades Raras (CIBERER), Av. Monforte de Lemos, 3-5, 28029, Madrid, Spain; Instituto de Biocomputación y Física de Sistemas Complejos (BIFI), Universidad de Zaragoza. C/ Mariano Esquillor (Edificio I+D), 50018, Zaragoza, Spain.
| | - Patricia Meade
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, C/ Miguel Servet, 177. 50013, Zaragoza, Spain and C/ Pedro Cerbuna, 12, 50009, Zaragoza, Spain; Instituto de Investigación Sanitaria (IIS) de Aragón, Av. San Juan Bosco, 13, 50009, Zaragoza, Spain; Centro de Investigaciones Biomédicas en Rd de Enfermedades Raras (CIBERER), Av. Monforte de Lemos, 3-5, 28029, Madrid, Spain; Instituto de Biocomputación y Física de Sistemas Complejos (BIFI), Universidad de Zaragoza. C/ Mariano Esquillor (Edificio I+D), 50018, Zaragoza, Spain.
| | - Eldris Iglesias
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, C/ Miguel Servet, 177. 50013, Zaragoza, Spain and C/ Pedro Cerbuna, 12, 50009, Zaragoza, Spain; Instituto de Investigación Sanitaria (IIS) de Aragón, Av. San Juan Bosco, 13, 50009, Zaragoza, Spain.
| | - Irene Jiménez-Salvador
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, C/ Miguel Servet, 177. 50013, Zaragoza, Spain and C/ Pedro Cerbuna, 12, 50009, Zaragoza, Spain; Instituto de Investigación Sanitaria (IIS) de Aragón, Av. San Juan Bosco, 13, 50009, Zaragoza, Spain.
| | - Julio Montoya
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, C/ Miguel Servet, 177. 50013, Zaragoza, Spain and C/ Pedro Cerbuna, 12, 50009, Zaragoza, Spain; Instituto de Investigación Sanitaria (IIS) de Aragón, Av. San Juan Bosco, 13, 50009, Zaragoza, Spain; Centro de Investigaciones Biomédicas en Rd de Enfermedades Raras (CIBERER), Av. Monforte de Lemos, 3-5, 28029, Madrid, Spain.
| | - Carmen Martínez-Cué
- Departamento de Fisiología y Farmacología. Facultad de Medicina, Universidad de Cantabria. Av. Herrera Oría, 39011, Santander, Spain.
| | - Eduardo Ruiz-Pesini
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, C/ Miguel Servet, 177. 50013, Zaragoza, Spain and C/ Pedro Cerbuna, 12, 50009, Zaragoza, Spain; Instituto de Investigación Sanitaria (IIS) de Aragón, Av. San Juan Bosco, 13, 50009, Zaragoza, Spain; Centro de Investigaciones Biomédicas en Rd de Enfermedades Raras (CIBERER), Av. Monforte de Lemos, 3-5, 28029, Madrid, Spain.
| |
Collapse
|
39
|
Szu J, Wojcinski A, Jiang P, Kesari S. Impact of the Olig Family on Neurodevelopmental Disorders. Front Neurosci 2021; 15:659601. [PMID: 33859549 PMCID: PMC8042229 DOI: 10.3389/fnins.2021.659601] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 03/08/2021] [Indexed: 12/13/2022] Open
Abstract
The Olig genes encode members of the basic helix-loop-helix (bHLH) family of transcription factors. Olig1, Olig2, and Olig3 are expressed in both the developing and mature central nervous system (CNS) and strictly regulate cellular specification and differentiation. Extensive studies have established functional roles of Olig1 and Olig2 in directing neuronal and glial formation during different stages in development. Recently, Olig2 overexpression was implicated in neurodevelopmental disorders down syndrome (DS) and autism spectrum disorder (ASD) but its influence on cognitive and intellectual defects remains unknown. In this review, we summarize the biological functions of the Olig family and how it uniquely promotes cellular diversity in the CNS. This is followed up with a discussion on how abnormal Olig2 expression impacts brain development and function in DS and ASD. Collectively, the studies described here emphasize vital features of the Olig members and their distinctive potential roles in neurodevelopmental disease states.
Collapse
Affiliation(s)
- Jenny Szu
- Department of Translational Neurosciences and Neurotherapeutics, Saint John's Cancer Institute, Providence Saint John's Health Center, Santa Monica, CA, United States
| | - Alexandre Wojcinski
- Department of Translational Neurosciences and Neurotherapeutics, Saint John's Cancer Institute, Providence Saint John's Health Center, Santa Monica, CA, United States
| | - Peng Jiang
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
| | - Santosh Kesari
- Department of Translational Neurosciences and Neurotherapeutics, Saint John's Cancer Institute, Providence Saint John's Health Center, Santa Monica, CA, United States.,Pacific Neuroscience Institute, Providence Saint John's Health Center, Santa Monica, CA, United States
| |
Collapse
|
40
|
Epigallocatechin-3-Gallate Plus Omega-3 Restores the Mitochondrial Complex I and F 0F 1-ATP Synthase Activities in PBMCs of Young Children with Down Syndrome: A Pilot Study of Safety and Efficacy. Antioxidants (Basel) 2021; 10:antiox10030469. [PMID: 33809669 PMCID: PMC8002266 DOI: 10.3390/antiox10030469] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/05/2021] [Accepted: 03/14/2021] [Indexed: 12/15/2022] Open
Abstract
Down syndrome (DS) is a major genetic cause of intellectual disability. DS pathogenesis has not been fully elucidated, and no specific pharmacological therapy is available. DYRK1A overexpression, oxidative stress and mitochondrial dysfunction were described in trisomy 21. Epigallocatechin-3-gallate (EGCG) is a multimodal nutraceutical with antioxidant properties. EGCG inhibits DYRK1A overexpression and corrects DS mitochondrial dysfunction in vitro. The present study explores safety profiles in DS children aged 1–8 years treated with EGCG (10 mg/kg/die, suspended in omega-3, per os, in fasting conditions, for 6 months) and EGCG efficacy in restoring mitochondrial complex I and F0F1-ATP synthase (complex V) deficiency, assessed on PBMCs. The Griffiths Mental Developmental Scales—Extended Revised (GMDS-ER) was used for developmental profiling. Results show that decaffeinated EGCG (>90%) plus omega-3 is safe in DS children and effective in reverting the deficit of mitochondrial complex I and V activities. Decline of plasma folates was observed in 21% of EGCG-treated patients and should be carefully monitored. GMDS-ER scores did not show differences between the treated group compared to the DS control group. In conclusion, EGCG plus omega-3 can be safely administered under medical supervision in DS children aged 1–8 years to normalize mitochondria respiratory chain complex activities, while results on the improvement of developmental performance are still inconclusive.
Collapse
|
41
|
Moreau M, Benhaddou S, Dard R, Tolu S, Hamzé R, Vialard F, Movassat J, Janel N. Metabolic Diseases and Down Syndrome: How Are They Linked Together? Biomedicines 2021; 9:biomedicines9020221. [PMID: 33671490 PMCID: PMC7926648 DOI: 10.3390/biomedicines9020221] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 02/15/2021] [Accepted: 02/17/2021] [Indexed: 12/13/2022] Open
Abstract
Down syndrome is a genetic disorder caused by the presence of a third copy of chromosome 21, associated with intellectual disabilities. Down syndrome is associated with anomalies of both the nervous and endocrine systems. Over the past decades, dramatic advances in Down syndrome research and treatment have helped to extend the life expectancy of these patients. Improved life expectancy is obviously a positive outcome, but it is accompanied with the need to address previously overlooked complications and comorbidities of Down syndrome, including obesity and diabetes, in order to improve the quality of life of Down syndrome patients. In this focused review, we describe the associations between Down syndrome and comorbidities, obesity and diabetes, and we discuss the understanding of proposed mechanisms for the association of Down syndrome with metabolic disorders. Drawing molecular mechanisms through which Type 1 diabetes and Type 2 diabetes could be linked to Down syndrome could allow identification of novel drug targets and provide therapeutic solutions to limit the development of metabolic and cognitive disorders.
Collapse
Affiliation(s)
- Manon Moreau
- Laboratoire Processus Dégénératifs, Université de Paris, BFA, UMR 8251, CNRS, Stress et Vieillissemen, F-75013 Paris, France; (M.M.); (S.B.); (R.D.)
| | - Soukaina Benhaddou
- Laboratoire Processus Dégénératifs, Université de Paris, BFA, UMR 8251, CNRS, Stress et Vieillissemen, F-75013 Paris, France; (M.M.); (S.B.); (R.D.)
| | - Rodolphe Dard
- Laboratoire Processus Dégénératifs, Université de Paris, BFA, UMR 8251, CNRS, Stress et Vieillissemen, F-75013 Paris, France; (M.M.); (S.B.); (R.D.)
- Genetics Deptartment, CHI Poissy St Germain-en-Laye, F-78300 Poissy, France;
- Université Paris-Saclay, UVSQ, INRAE, ENVA, BREED, F-78350 Jouy-en-Josas, France
| | - Stefania Tolu
- Laboratoire de Biologie et Pathologie du Pancréas Endocrine, Université de Paris, BFA, UMR 8251, CNRS, F-75013 Paris, France; (S.T.); (R.H.); (J.M.)
| | - Rim Hamzé
- Laboratoire de Biologie et Pathologie du Pancréas Endocrine, Université de Paris, BFA, UMR 8251, CNRS, F-75013 Paris, France; (S.T.); (R.H.); (J.M.)
| | - François Vialard
- Genetics Deptartment, CHI Poissy St Germain-en-Laye, F-78300 Poissy, France;
- Université Paris-Saclay, UVSQ, INRAE, ENVA, BREED, F-78350 Jouy-en-Josas, France
| | - Jamileh Movassat
- Laboratoire de Biologie et Pathologie du Pancréas Endocrine, Université de Paris, BFA, UMR 8251, CNRS, F-75013 Paris, France; (S.T.); (R.H.); (J.M.)
| | - Nathalie Janel
- Laboratoire Processus Dégénératifs, Université de Paris, BFA, UMR 8251, CNRS, Stress et Vieillissemen, F-75013 Paris, France; (M.M.); (S.B.); (R.D.)
- Correspondence: ; Tel.: +33-1-57-27-83-60; Fax: +33-1-57-27-83-54
| |
Collapse
|
42
|
Panisi C, Guerini FR, Abruzzo PM, Balzola F, Biava PM, Bolotta A, Brunero M, Burgio E, Chiara A, Clerici M, Croce L, Ferreri C, Giovannini N, Ghezzo A, Grossi E, Keller R, Manzotti A, Marini M, Migliore L, Moderato L, Moscone D, Mussap M, Parmeggiani A, Pasin V, Perotti M, Piras C, Saresella M, Stoccoro A, Toso T, Vacca RA, Vagni D, Vendemmia S, Villa L, Politi P, Fanos V. Autism Spectrum Disorder from the Womb to Adulthood: Suggestions for a Paradigm Shift. J Pers Med 2021; 11:70. [PMID: 33504019 PMCID: PMC7912683 DOI: 10.3390/jpm11020070] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 01/10/2021] [Accepted: 01/19/2021] [Indexed: 02/07/2023] Open
Abstract
The wide spectrum of unique needs and strengths of Autism Spectrum Disorders (ASD) is a challenge for the worldwide healthcare system. With the plethora of information from research, a common thread is required to conceptualize an exhaustive pathogenetic paradigm. The epidemiological and clinical findings in ASD cannot be explained by the traditional linear genetic model, hence the need to move towards a more fluid conception, integrating genetics, environment, and epigenetics as a whole. The embryo-fetal period and the first two years of life (the so-called 'First 1000 Days') are the crucial time window for neurodevelopment. In particular, the interplay and the vicious loop between immune activation, gut dysbiosis, and mitochondrial impairment/oxidative stress significantly affects neurodevelopment during pregnancy and undermines the health of ASD people throughout life. Consequently, the most effective intervention in ASD is expected by primary prevention aimed at pregnancy and at early control of the main effector molecular pathways. We will reason here on a comprehensive and exhaustive pathogenetic paradigm in ASD, viewed not just as a theoretical issue, but as a tool to provide suggestions for effective preventive strategies and personalized, dynamic (from womb to adulthood), systemic, and interdisciplinary healthcare approach.
Collapse
Affiliation(s)
- Cristina Panisi
- Fondazione Istituto Sacra Famiglia ONLUS, Cesano Boscone, 20090 Milan, Italy;
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy;
| | - Franca Rosa Guerini
- IRCCS Fondazione Don Carlo Gnocchi, ONLUS, 20148 Milan, Italy; (M.C.); (M.S.)
| | | | - Federico Balzola
- Division of Gastroenterology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, University of Turin, 10126 Turin, Italy;
| | - Pier Mario Biava
- Scientific Institute of Research and Care Multimedica, 20138 Milan, Italy;
| | - Alessandra Bolotta
- DIMES, School of Medicine, University of Bologna, 40126 Bologna, Italy; (P.M.A.); (A.B.); (A.G.)
| | - Marco Brunero
- Department of Pediatric Surgery, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| | - Ernesto Burgio
- ECERI—European Cancer and Environment Research Institute, Square de Meeus 38-40, 1000 Bruxelles, Belgium;
| | - Alberto Chiara
- Dipartimento Materno Infantile ASST, 27100 Pavia, Italy;
| | - Mario Clerici
- IRCCS Fondazione Don Carlo Gnocchi, ONLUS, 20148 Milan, Italy; (M.C.); (M.S.)
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy
| | - Luigi Croce
- Centro Domino per l’Autismo, Universita’ Cattolica Brescia, 20139 Milan, Italy;
| | - Carla Ferreri
- National Research Council of Italy, Institute of Organic Synthesis and Photoreactivity (ISOF), 40129 Bologna, Italy;
| | - Niccolò Giovannini
- Department of Obstetrics and Gynecology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Alessandro Ghezzo
- DIMES, School of Medicine, University of Bologna, 40126 Bologna, Italy; (P.M.A.); (A.B.); (A.G.)
| | - Enzo Grossi
- Autism Research Unit, Villa Santa Maria Foundation, 22038 Tavernerio, Italy;
| | - Roberto Keller
- Adult Autism Centre DSM ASL Città di Torino, 10138 Turin, Italy;
| | - Andrea Manzotti
- RAISE Lab, Foundation COME Collaboration, 65121 Pescara, Italy;
| | - Marina Marini
- DIMES, School of Medicine, University of Bologna, 40126 Bologna, Italy; (P.M.A.); (A.B.); (A.G.)
| | - Lucia Migliore
- Medical Genetics Laboratories, Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy; (L.M.); (A.S.)
| | - Lucio Moderato
- Fondazione Istituto Sacra Famiglia ONLUS, Cesano Boscone, 20090 Milan, Italy;
| | - Davide Moscone
- Associazione Spazio Asperger ONLUS, Centro Clinico CuoreMenteLab, 00141 Rome, Italy;
| | - Michele Mussap
- Neonatal Intensive Care Unit, Department of Surgical Sciences, Puericulture Institute and Neonatal Section, Azienda Ospedaliera Universitaria, 09100 Cagliari, Italy; (M.M.); (V.F.)
| | - Antonia Parmeggiani
- Child Neurology and Psychiatry Unit, IRCCS ISNB, S. Orsola-Malpighi Hospital, Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy;
| | - Valentina Pasin
- Milan Institute for health Care and Advanced Learning, 20124 Milano, Italy;
| | | | - Cristina Piras
- Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy;
| | - Marina Saresella
- IRCCS Fondazione Don Carlo Gnocchi, ONLUS, 20148 Milan, Italy; (M.C.); (M.S.)
| | - Andrea Stoccoro
- Medical Genetics Laboratories, Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy; (L.M.); (A.S.)
| | - Tiziana Toso
- Unione Italiana Lotta alla Distrofia Muscolare UILDM, 35100 Padova, Italy;
| | - Rosa Anna Vacca
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council of Italy, 70126 Bari, Italy;
| | - David Vagni
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy, 98164 Messina, Italy;
| | | | - Laura Villa
- Scientific Institute, IRCCS Eugenio Medea, Via Don Luigi Monza 20, 23842 Bosisio Parini, Italy;
| | - Pierluigi Politi
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy;
| | - Vassilios Fanos
- Neonatal Intensive Care Unit, Department of Surgical Sciences, Puericulture Institute and Neonatal Section, Azienda Ospedaliera Universitaria, 09100 Cagliari, Italy; (M.M.); (V.F.)
- Neonatal Intensive Care Unit, Azienda Ospedaliera Universitaria, 09042 Cagliari, Italy
| |
Collapse
|
43
|
Uguagliati B, Al-Absi AR, Stagni F, Emili M, Giacomini A, Guidi S, Nyengaard JR, Bartesaghi R. Early appearance of developmental alterations in the dendritic tree of the hippocampal granule cells in the Ts65Dn model of Down syndrome. Hippocampus 2021; 31:435-447. [PMID: 33464704 DOI: 10.1002/hipo.23303] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/22/2020] [Accepted: 01/09/2021] [Indexed: 12/28/2022]
Abstract
Down syndrome (DS), a genetic condition caused by triplication of chromosome 21, is characterized by alterations in various cognitive domains, including hippocampus-dependent memory functions, starting from early life stages. The major causes of intellectual disability in DS are prenatal neurogenesis alterations followed by impairment of dendritic development in early infancy. While there is evidence that the Ts65Dn mouse, the most widely used model of DS, exhibits dendritic alterations in adulthood, no studies are available regarding the onset of dendritic pathology. The goal of the current study was to establish whether this model exhibits early dendritic alterations in the hippocampus, a region whose function is severely damaged in DS. To this purpose, in Golgi-stained brains, we evaluated the dendritic arborization and dendritic spines of the granule cells of the hippocampal dentate gyrus in Ts65Dn mice aged 8 (P8) and 15 (P15) days. While P15 Ts65Dn mice exhibited a notably hypotrophic dendritic arbor and a reduced spine density, P8 mice exhibited a moderate reduction in the number of dendritic ramifications and no differences in spine density in comparison with their euploid counterparts. Both in P8 and P15 mice, spines were longer and had a longer neck, suggesting possible alterations in synaptic function. Moreover, P8 and P15 Ts65Dn mice had more thin spines and fewer stubby spines in comparison with euploid mice. Our study provides novel evidence on the onset of dendritic pathology, one of the causes of intellectual disability in DS, showing that it is already detectable in the dentate gyrus of Ts65Dn pups. This evidence strengthens the suitability of this model of DS as a tool to study dendritic pathology in DS and to test the efficacy of early therapeutic interventions aimed at ameliorating hippocampal development and, therefore, memory functions in children with DS.
Collapse
Affiliation(s)
- Beatrice Uguagliati
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Abdel-Rahman Al-Absi
- Core Centre for Molecular Morphology, Section for Stereology and Microscopy, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Fiorenza Stagni
- Department for Life Quality Studies, University of Bologna, Rimini, Italy
| | - Marco Emili
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Andrea Giacomini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Sandra Guidi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Jens Randel Nyengaard
- Core Centre for Molecular Morphology, Section for Stereology and Microscopy, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Renata Bartesaghi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
44
|
Tewari D, Sah AN, Bawari S, Nabavi SF, Dehpour AR, Shirooie S, Braidy N, Fiebich BL, Vacca RA, Nabavi SM. Role of Nitric Oxide in Neurodegeneration: Function, Regulation, and Inhibition. Curr Neuropharmacol 2020; 19:114-126. [PMID: 32348225 PMCID: PMC8033982 DOI: 10.2174/1570159x18666200429001549] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 03/17/2020] [Accepted: 04/24/2020] [Indexed: 12/14/2022] Open
Abstract
Reactive nitrogen species (RNS) and reactive oxygen species (ROS), collectively known as reactive oxygen and nitrogen species (RONS), are the products of normal cellular metabolism and interact with several vital biomolecules including nucleic acid, proteins, and membrane lipids and alter their function in an irreversible manner which can lead to cell death. There is an imperative role for oxidative stress in the pathogenesis of cognitive impairments and the development and progression of neural injury. Elevated production of higher amounts of nitric oxide (NO) takes place in numerous pathological conditions, such as neurodegenerative diseases, inflammation, and ischemia, which occur concurrently with elevated nitrosative/oxidative stress. The enzyme nitric oxide synthase (NOS) is responsible for the generation of NO in different cells by conversion of L-arginine (Arg) to L-citrulline. Therefore, the NO signaling pathway represents a viable therapeutic target. Naturally occurring polyphenols targeting the NO signaling pathway can be of major importance in the field of neurodegeneration and related complications. Here, we comprehensively review the importance of NO and its production in the human body and afterwards highlight the importance of various natural products along with their mechanisms against various neurodegenerative diseases involving their effect on NO production.
Collapse
Affiliation(s)
- Devesh Tewari
- Department of Pharmacognosy, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Archana N Sah
- Department of Pharmaceutical Sciences, Faculty of Technology, Bhimtal Campus, Kumaun University, Nainital, Uttarakhand 263136, India
| | - Sweta Bawari
- School of Pharmacy, Sharda University, Knowledge Park-III, Greater Noida, Uttar Pradesh, 201310, India
| | - Seyed F Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran 1435916471, Iran
| | - Ahmad R Dehpour
- Department of Pharmacology, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Samira Shirooie
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Nady Braidy
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Australia
| | - Bernd L Fiebich
- Neuroimmunology and Neurochemistry Research Group, Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Rosa A Vacca
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Council of Research, Bari, Italy
| | - Seyed M Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran 1435916471, Iran
| |
Collapse
|
45
|
Pecze L, Randi EB, Szabo C. Meta-analysis of metabolites involved in bioenergetic pathways reveals a pseudohypoxic state in Down syndrome. Mol Med 2020; 26:102. [PMID: 33167881 PMCID: PMC7653803 DOI: 10.1186/s10020-020-00225-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 10/12/2020] [Indexed: 12/14/2022] Open
Abstract
Clinical observations and preclinical studies both suggest that Down syndrome (DS) may be associated with significant metabolic and bioenergetic alterations. However, the relevant scientific literature has not yet been systematically reviewed. The aim of the current study was to conduct a meta-analysis of metabolites involved in bioenergetics pathways in DS to conclusively determine the difference between DS and control subjects. We discuss these findings and their potential relevance in the context of pathogenesis and experimental therapy of DS. Articles published before July 1, 2020, were identified by using the search terms “Down syndrome” and “metabolite name” or “trisomy 21” and “metabolite name”. Moreover, DS-related metabolomics studies and bioenergetics literature were also reviewed. 41 published reports and associated databases were identified, from which the descriptive information and the relevant metabolomic parameters were extracted and analyzed. Mixed effect model revealed the following changes in DS: significantly decreased ATP, CoQ10, homocysteine, serine, arginine and tyrosine; slightly decreased ADP; significantly increased uric acid, succinate, lactate and cysteine; slightly increased phosphate, pyruvate and citrate. However, the concentrations of AMP, 2,3-diphosphoglycerate, glucose, and glutamine were comparable in the DS vs. control populations. We conclude that cells of subjects with DS are in a pseudo-hypoxic state: the cellular metabolic and bio-energetic mechanisms exhibit pathophysiological alterations that resemble the cellular responses associated with hypoxia, even though the supply of the cells with oxygen is not disrupted. This fundamental alteration may be, at least in part, responsible for a variety of functional deficits associated with DS, including reduced exercise difference, impaired neurocognitive status and neurodegeneration.
Collapse
Affiliation(s)
- Laszlo Pecze
- Chair of Pharmacology, Section of Medicine, University of Fribourg, Fribourg, Switzerland
| | - Elisa B Randi
- Chair of Pharmacology, Section of Medicine, University of Fribourg, Fribourg, Switzerland
| | - Csaba Szabo
- Chair of Pharmacology, Section of Medicine, University of Fribourg, Fribourg, Switzerland.
| |
Collapse
|
46
|
Mansur F, Alarcon JM, Stackpole EE, Wang R, Richter JD. Noncanonical cytoplasmic poly(A) polymerases regulate RNA levels, alternative RNA processing, and synaptic plasticity but not hippocampal-dependent behaviours. RNA Biol 2020; 18:962-971. [PMID: 32954964 DOI: 10.1080/15476286.2020.1824061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Noncanonical poly(A) polymerases are frequently tethered to mRNA 3' untranslated regions and regulate poly(A) tail length and resulting translation. In the brain, one such poly(A) polymerase is Gld2, which is anchored to mRNA by the RNA-binding protein CPEB1 to control local translation at postsynaptic regions. Depletion of CPEB1 or Gld2 from the mouse hippocampus results in a deficit in long-term potentiation (LTP), but only depletion of CPEB1 alters animal behaviour. To test whether a related enzyme, Gld4, compensates for the lack of Gld2, we separately or simultaneously depleted both proteins from hippocampal area CA1 and again found little change in animal behaviour, but observed a deficit in LTP as well as an increase in long-term depression (LTD), two forms of protein synthesis-dependent synaptic plasticity. RNA-seq data from Gld2, Gld4, and Gld2/Gld4-depleted hippocampus show widespread changes in steady state RNA levels, alternative splicing, and alternative poly(A) site selection. Many of the RNAs subject to these alterations encode proteins that mediate synaptic function, suggesting a molecular foundation for impaired synaptic plasticity.
Collapse
Affiliation(s)
- Fernanda Mansur
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Juan Marcos Alarcon
- Department of Pathology, SUNY Downstate Health Sciences University, Brooklyn, New York, USA
| | - Emily E Stackpole
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Ruijia Wang
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Joel D Richter
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
47
|
Bonucci M, Raggi R, Vacca RA. Polydatin and its potential protective effect on COVID-19. Clin Nutr 2020; 39:3850-3851. [PMID: 33097304 PMCID: PMC7543899 DOI: 10.1016/j.clnu.2020.09.052] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 09/01/2020] [Indexed: 01/19/2023]
Affiliation(s)
- Massimo Bonucci
- Association for Research on Integrative Oncological Therapies (ARTOI), Rome, Italy.
| | | | - Rosa Anna Vacca
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Council of Research, Bari, Italy.
| |
Collapse
|
48
|
Chung J, Donelan K, Macklin EA, Schwartz A, Elsharkawi I, Torres A, Hsieh YG, Parker H, Lorenz S, Patsiogiannis V, Santoro SL, Wylie M, Clarke L, Estey G, Baker S, Bauer PE, Bull M, Chicoine B, Cullen S, Frey-Vogel A, Gallagher M, Hasan R, Lamb A, Majewski L, Mast J, Riddell T, Sepucha K, Skavlem M, Skotko BG. A randomized controlled trial of an online health tool about Down syndrome. Genet Med 2020; 23:163-173. [PMID: 32879436 DOI: 10.1038/s41436-020-00952-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/13/2020] [Accepted: 08/13/2020] [Indexed: 12/22/2022] Open
Abstract
PURPOSE We sought to determine if a novel online health tool, called Down Syndrome Clinic to You (DSC2U), could improve adherence to national Down syndrome (DS) guidelines. We also sought to determine if primary care providers (PCPs) and caregivers are satisfied with this personalized online health tool. METHODS In a national, randomized controlled trial of 230 caregivers who had children or dependents with DS without access to a DS specialist, 117 were randomized to receive DSC2U and 113 to receive usual care. The primary outcome was adherence to five health evaluations indicated by national guidelines for DS. DSC2U is completed electronically, in all mobile settings, by caregivers at home. The outputs-personalized checklists-are used during annual wellness visits with the patient's PCP. RESULTS A total of 213 participants completed a 7-month follow-up evaluation. In the intention-to-treat analysis, the intervention group had a 1.6-fold increase in the number of indicated evaluations that were recommended by the primary care provider or completed compared with controls. Both caregivers and PCPs reported high levels of satisfaction with DSC2U. CONCLUSIONS DSC2U improved adherence to the national DS health-care guidelines with a novel modality that was highly valued by both caregivers and PCPs.
Collapse
Affiliation(s)
- Jeanhee Chung
- Laboratory of Computer Science, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Karen Donelan
- Department of Medicine, Harvard Medical School, Boston, MA, USA.,Health Policy Research Center, Mongan Institute, Massachusetts General Hospital, Boston, MA, USA
| | - Eric A Macklin
- Biostatistics Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Alison Schwartz
- Down Syndrome Program, Division of Medical Genetics and Metabolism, Department of Pediatrics, Massachusetts General Hospital, Boston, MA, USA.,Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Ibrahim Elsharkawi
- Down Syndrome Program, Division of Medical Genetics and Metabolism, Department of Pediatrics, Massachusetts General Hospital, Boston, MA, USA
| | - Amy Torres
- Down Syndrome Program, Division of Medical Genetics and Metabolism, Department of Pediatrics, Massachusetts General Hospital, Boston, MA, USA
| | - Yichuan Grace Hsieh
- Laboratory of Computer Science, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Holly Parker
- Laboratory of Computer Science, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Stephen Lorenz
- Laboratory of Computer Science, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Vasiliki Patsiogiannis
- Down Syndrome Program, Division of Medical Genetics and Metabolism, Department of Pediatrics, Massachusetts General Hospital, Boston, MA, USA
| | - Stephanie L Santoro
- Down Syndrome Program, Division of Medical Genetics and Metabolism, Department of Pediatrics, Massachusetts General Hospital, Boston, MA, USA.,Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Mark Wylie
- Laboratory of Computer Science, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Lloyd Clarke
- Laboratory of Computer Science, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Greg Estey
- Laboratory of Computer Science, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sandra Baker
- Down Syndrome Association of Los Angeles, Los Angeles, CA, USA
| | | | - Marilyn Bull
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Brian Chicoine
- Advocate Medical Group Adult Down Syndrome Center, Park Ridge, IL, USA
| | - Sarah Cullen
- Massachusetts Down Syndrome Congress, Boston, MA, USA
| | - Ariel Frey-Vogel
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | | | - Reem Hasan
- Department of Internal Medicine and Pediatrics, Oregon Health & Science University, Portland, OR, USA
| | - Ashley Lamb
- Health and Wellness, University of New Hampshire, Durham, NH, USA
| | | | | | - Travis Riddell
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | - Karen Sepucha
- Health Decision Sciences Center, Massachusetts General Hospital, Boston, MA, USA
| | | | - Brian G Skotko
- Down Syndrome Program, Division of Medical Genetics and Metabolism, Department of Pediatrics, Massachusetts General Hospital, Boston, MA, USA. .,Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
49
|
Rueda N, Vidal V, García-Cerro S, Puente A, Campa V, Lantigua S, Narcís O, Bartesaghi R, Martínez-Cué C. Prenatal, but not Postnatal, Curcumin Administration Rescues Neuromorphological and Cognitive Alterations in Ts65Dn Down Syndrome Mice. J Nutr 2020; 150:2478-2489. [PMID: 32729926 DOI: 10.1093/jn/nxaa207] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/27/2020] [Accepted: 06/26/2020] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND The cognitive dysfunction in Down syndrome (DS) is partially caused by deficient neurogenesis during fetal stages. Curcumin enhances neurogenesis and learning and memory. OBJECTIVES We aimed to test the ability of curcumin to rescue the neuromorphological and cognitive alterations of the Ts65Dn (TS) mouse model of DS when administered prenatally or during early postnatal stages, and to evaluate whether these effects were maintained several weeks after the treatment. METHODS To evaluate the effects of prenatal curcumin administration, 65 pregnant TS females were subcutaneously treated with curcumin (300 mg/kg) or vehicle from ED (Embryonic Day) 10 to PD (Postnatal Day) 2. All the analyses were performed on their TS and Control (CO) male and female progeny. At PD2, the changes in neurogenesis, cellularity, and brain weight were analyzed in 30 TS and CO pups. The long-term effects of prenatal curcumin were evaluated in another cohort of 44 TS and CO mice between PD30 and PD45. The neuromorphological effects of the early postnatal administration of curcumin were assessed on PD15 in 30 male and female TS and CO pups treated with curcumin (300 mg/kg) or vehicle from PD2 to PD15. The long-term neuromorphological and cognitive effects were assessed from PD60 to PD90 in 45 mice. Data was compared by ANOVAs. RESULTS Prenatal administration of curcumin increased the brain weight (+45%, P < 0.001), the density of BrdU (bromodeoxyuridine)- (+150%, P < 0.001) and DAPI (4',6-diamidino-2-phenylindole)- (+38%, P = 0.005) positive cells, and produced a long-term improvement of cognition in TS (+35%, P = 0.007) mice with respect to vehicle-treated mice. Postnatal administration of curcumin did not rescue any of the short- or long-term altered phenotypes of TS mice. CONCLUSION The beneficial effects of prenatal curcumin administration to TS mice suggest that it could be a therapeutic strategy to treat DS cognitive disabilities.
Collapse
Affiliation(s)
- Noemí Rueda
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Verónica Vidal
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Susana García-Cerro
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Alba Puente
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Víctor Campa
- Institute of Molecular Biology and Biomedicine, Santander, Cantabria, Spain
| | - Sara Lantigua
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Oriol Narcís
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Renata Bartesaghi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Carmen Martínez-Cué
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| |
Collapse
|
50
|
4-Phenylbutyrate ameliorates apoptotic neural cell death in Down syndrome by reducing protein aggregates. Sci Rep 2020; 10:14047. [PMID: 32820178 PMCID: PMC7441064 DOI: 10.1038/s41598-020-70362-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 07/27/2020] [Indexed: 01/11/2023] Open
Abstract
Individuals with Down syndrome (DS) commonly show unique pathological phenotypes throughout their life span. Besides the specific effects of dosage-sensitive genes on chromosome 21, recent studies have demonstrated that the gain of a chromosome exerts an adverse impact on cell physiology, regardless of the karyotype. Although dysregulated transcription and perturbed protein homeostasis are observed in common in human fibroblasts with trisomy 21, 18, and 13, whether and how this aneuploidy-associated stress acts on other cell lineages and affects the pathophysiology are unknown. Here, we investigated cellular stress responses in human trisomy 21 and 13 neurons differentiated from patient-derived induced pluripotent stem cells. Neurons of both trisomies showed increased vulnerability to apoptotic cell death, accompanied by dysregulated protein homeostasis and upregulation of the endoplasmic reticulum stress pathway. In addition, misfolded protein aggregates, comprising various types of neurodegenerative disease-related proteins, were abnormally accumulated in trisomic neurons. Intriguingly, treatment with sodium 4-phenylbutyrate, a chemical chaperone, successfully decreased the formation of protein aggregates and prevented the progression of cell apoptosis in trisomic neurons. These results suggest that aneuploidy-associated stress might be a therapeutic target for the neurodegenerative phenotypes in DS.
Collapse
|