1
|
Feng Y, Yang X, Hou Y, Zhou Y, Leverenz JB, Eng C, Pieper AA, Goate A, Shen Y, Cheng F. Widespread transposable element dysregulation in human aging brains with Alzheimer's disease. Alzheimers Dement 2024; 20:7495-7517. [PMID: 39356058 PMCID: PMC11567813 DOI: 10.1002/alz.14164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 07/08/2024] [Accepted: 07/11/2024] [Indexed: 10/03/2024]
Abstract
INTRODUCTION Transposable element (TE) dysregulation is associated with neuroinflammation in Alzheimer's disease (AD) brains. Yet, TE quantitative trait loci (teQTL) have not been well characterized in human aged brains with AD. METHODS We leveraged large-scale bulk and single-cell RNA sequencing, whole-genome sequencing (WGS), and xQTL from three human AD brain biobanks to characterize TE expression dysregulation and experimentally validate AD-associated TEs using CRISPR interference (CRISPRi) assays in human induced pluripotent stem cell (iPSC)-derived neurons. RESULTS We identified 26,188 genome-wide significant TE expression QTLs (teQTLs) in human aged brains. Subsequent colocalization analysis of teQTLs with AD genetic loci identified AD-associated teQTLs and linked locus TEs. Using CRISPRi assays, we pinpointed a neuron-specific suppressive role of the activated short interspersed nuclear element (SINE; chr11:47608036-47608220) on expression of C1QTNF4 via reducing neuroinflammation in human iPSC-derived neurons. DISCUSSION We identified widespread TE dysregulation in human AD brains and teQTLs offer a complementary analytic approach to identify likely AD risk genes. HIGHLIGHTS Widespread transposable element (TE) dysregulations are observed in human aging brains with degrees of neuropathology, apolipoprotein E (APOE) genotypes, and neuroinflammation in Alzheimer's disease (AD). A catalog of TE quantitative trait loci (teQTLs) in human aging brains was created using matched RNA sequencing and whole-genome sequencing data. CRISPR interference assays reveal that an upregulated intergenic TE from the MIR family (chr11: 47608036-47608220) suppresses expression of its nearest anti-inflammatory gene C1QTNF4 in human induced pluripotent stem cell-derived neurons.
Collapse
Affiliation(s)
- Yayan Feng
- Cleveland Clinic Genome CenterLerner Research InstituteCleveland ClinicClevelandOhioUSA
- Genomic Medicine InstituteLerner Research InstituteCleveland ClinicClevelandOhioUSA
| | - Xiaoyu Yang
- Cleveland Clinic Genome CenterLerner Research InstituteCleveland ClinicClevelandOhioUSA
- Institute for Human GeneticsUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Yuan Hou
- Cleveland Clinic Genome CenterLerner Research InstituteCleveland ClinicClevelandOhioUSA
- Genomic Medicine InstituteLerner Research InstituteCleveland ClinicClevelandOhioUSA
| | - Yadi Zhou
- Cleveland Clinic Genome CenterLerner Research InstituteCleveland ClinicClevelandOhioUSA
- Genomic Medicine InstituteLerner Research InstituteCleveland ClinicClevelandOhioUSA
| | - James B. Leverenz
- Lou Ruvo Center for Brain HealthNeurological InstituteCleveland ClinicClevelandOhioUSA
| | - Charis Eng
- Genomic Medicine InstituteLerner Research InstituteCleveland ClinicClevelandOhioUSA
- Department of Molecular MedicineCleveland Clinic Lerner College of MedicineCase Western Reserve UniversityClevelandOhioUSA
- Department of Genetics and Genome SciencesCase Western Reserve University School of MedicineClevelandOhioUSA
- Case Comprehensive Cancer CenterCase Western Reserve University School of MedicineClevelandOhioUSA
| | - Andrew A. Pieper
- Department of PsychiatryCase Western Reserve UniversityClevelandOhioUSA
- Brain Health Medicines CenterHarrington Discovery InstituteUniversity Hospitals Cleveland Medical CenterClevelandOhioUSA
- Geriatric PsychiatryGRECCLouis Stokes Cleveland VA Medical CenterClevelandOhioUSA
- Institute for Transformative Molecular MedicineSchool of MedicineCase Western Reserve UniversityClevelandOhioUSA
- Department of NeurosciencesCase Western Reserve UniversitySchool of MedicineClevelandOhioUSA
- Department of PathologyCase Western Reserve UniversitySchool of MedicineClevelandOhioUSA
| | - Alison Goate
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Nash Department of NeuroscienceIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Ronald M. Loeb Center for Alzheimer's DiseaseIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Yin Shen
- Institute for Human GeneticsUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
- Department of NeurologyUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
- Weill Institute for NeurosciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Feixiong Cheng
- Cleveland Clinic Genome CenterLerner Research InstituteCleveland ClinicClevelandOhioUSA
- Genomic Medicine InstituteLerner Research InstituteCleveland ClinicClevelandOhioUSA
- Department of Molecular MedicineCleveland Clinic Lerner College of MedicineCase Western Reserve UniversityClevelandOhioUSA
- Case Comprehensive Cancer CenterCase Western Reserve University School of MedicineClevelandOhioUSA
| |
Collapse
|
2
|
Allerton TD, Stampley JE, Li Z, Yu X, Quiariate H, Doiron JE, White G, Wigger Z, Gartia MR, Lefer DJ, Soto P, Irving BA. Nitric oxide donors rescue metabolic and mitochondrial dysfunction in obese Alzheimer's model. Sci Rep 2024; 14:26118. [PMID: 39478095 PMCID: PMC11525932 DOI: 10.1038/s41598-024-75870-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 10/08/2024] [Indexed: 11/02/2024] Open
Abstract
Reduced nitric oxide (NO) bioavailability is a pathological link between obesity and Alzheimer's disease (AD). Obesity-associated metabolic and mitochondrial bioenergetic dysfunction are key drivers of AD pathology. The hypothalamus is a critical brain region during the development of obesity and dysfunction is an area implicated in the development of AD. NO is an essential mediator of blood flow and mitochondrial bioenergetic function, but the role of NO in obesity-AD is not entirely clear. We investigated diet-induced obesity in female APPswe/PS1dE9 (APP) mouse model of AD, which we treated with two different NO donors (sodium nitrite or L-citrulline). After 26 weeks of a high-fat diet, female APP mice had higher adiposity, insulin resistance, and mitochondrial dysfunction (hypothalamus) than non-transgenic littermate (wild type) controls. Treatment with either sodium nitrite or L-citrulline did not reduce adiposity but improved whole-body energy expenditure, substrate oxidation, and insulin sensitivity. Notably, both NO donors restored hypothalamic mitochondrial respiration in APP mice. Our findings suggest that NO is an essential mediator of whole-body metabolism and hypothalamic mitochondrial function, which are severely impacted by the dual insults of obesity and AD pathology.
Collapse
Affiliation(s)
- Timothy D Allerton
- Vascular Metabolism Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA.
| | - James E Stampley
- Department of Kinesiology, Louisiana State University, Baton Rouge, LA, USA
| | - Zhen Li
- Department of Cardiac Surgery, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Xiaoman Yu
- Department of Cardiac Surgery, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Heather Quiariate
- Vascular Metabolism Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
- Department of Kinesiology, Louisiana State University, Baton Rouge, LA, USA
| | - Jake E Doiron
- Vascular Metabolism Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - Ginger White
- Department of Psychology, Louisiana State University, Baton Rouge, LA, USA
| | - Zach Wigger
- Adipocyte Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Manas Ranjan Gartia
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA, USA
| | - David J Lefer
- Department of Cardiac Surgery, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Paul Soto
- Department of Psychology, Louisiana State University, Baton Rouge, LA, USA
| | - Brian A Irving
- Department of Kinesiology, Louisiana State University, Baton Rouge, LA, USA
| |
Collapse
|
3
|
Zhu H, Li X, Wang J, Wang H, Zhao S, Tian Y, Su Y. Transcriptomic analysis reveals differentially expressed genes associated with meat quality in Chinese Dagu chicken and AA + broiler roosters. BMC Genomics 2024; 25:1002. [PMID: 39455924 PMCID: PMC11515088 DOI: 10.1186/s12864-024-10927-6] [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: 02/26/2024] [Accepted: 10/21/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND With the improvement of living standards, the quality of chicken has become a significant concern. Chinese Dagu Chicken (dual-purpose type) and Arbor Acres plus broiler (AA+ broiler) (meat-type) were selected as the research subjects in this study, the meat quality of the breast and leg muscles were measured. However, the molecular mechanism(s) underlying regulation of muscle development are not yet fully elucidated. Therefore, finding molecular markers or major genes that regulate muscle quality has become a crucial breakthrough in chicken breeding. Unraveling the molecular mechanism behind meat traits in chicken and other domestic fowl is facilitated by identifying the key genes associated with these developmental events. Here, a comparative transcriptomic analysis of chicken meat was conducted on breast muscles (BM) and leg muscles (LM) in AA+ broilers (AA) and Dagu chicken (DG) to explore the differences in their meat traits employing RNA-seq. RESULTS Twelve cDNA libraries of BM and LM from AA and DG were constructed from four experimental groups, yielding 14,464 genes. Among them, Dagu chicken breast muscles (DGB) vs AA+ broilers breast muscles (AAB) showed 415 upregulated genes and 449 downregulated genes, Dagu chicken leg muscles (DGL) vs AA+ broilers leg muscles (AAL) exhibited 237 upregulated genes and 278 downregulated genes, DGL vs DGB demonstrated 391 upregulated genes and 594 downregulated genes, and AAL vs AAB displayed 122 upregulated genes and 154 downregulated genes. 13 genes, including nine upregulated genes (COX5A, COX7C, NDUFV1, UQCRFS1, UQCR11, BRT-1, FGF14, TMOD1, MYOZ2) and four downregulated genes (MYBPC3, MYO7B, MTMR7, and TNNC1), were found to be associated with the oxidative phosphorylation signaling pathway. Further analysis revealed that the differentially expressed genes (DEGs) from muscle were enriched in various pathways, such as metabolic pathways, oxidative phosphorylation, carbon metabolism, glycolysis, extracellular matrix-receptor interaction, biosynthesis of amino acids, focal adhesion, vascular smooth muscle contraction, and cardiac muscle contraction, all of which are involved in muscle development and metabolism. This study also measured the meat quality of the breast and leg muscles from the two breeds, which demonstrated superior overall meat quality in Chinese Dagu Chicken compared to the AA+ broiler. CONCLUSIONS Our findings show that the meat quality of dual-purpose breeds (Chinese Dagu chicken) is higher than meat-type (AA+ broiler), which may be related to the DEGs regulating muscle development and metabolism. Our findings also provide transcriptomic insights for a comparative analysis of molecular mechanisms underlying muscle development between the two breeds, and have practical implications for the improvement of chicken breeding practices.
Collapse
Affiliation(s)
- Hongyan Zhu
- College of Basic Medical Science, Jinzhou Medical University, Jinzhou , Liaoning, 121001, China
- Key Laboratory of Molecular Cell Biology and New Drug Development of the Education, Department of Liaoning Province, Jinzhou, Liaoning, 121001, China
| | - Xiaohan Li
- College of Animal Husbandry and Veterinary, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
| | - Jie Wang
- College of Animal Husbandry and Veterinary, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
| | - Haoming Wang
- College of Basic Medical Science, Jinzhou Medical University, Jinzhou , Liaoning, 121001, China
| | - Song Zhao
- Key Laboratory of Molecular Cell Biology and New Drug Development of the Education, Department of Liaoning Province, Jinzhou, Liaoning, 121001, China
- College of Life Science Institute, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
| | - Yumin Tian
- College of Animal Husbandry and Veterinary, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
| | - Yuhong Su
- College of Food and Health, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China.
| |
Collapse
|
4
|
Yang S, Tian M, Dai Y, Wang R, Yamada S, Feng S, Wang Y, Chhangani D, Ou T, Li W, Guo X, McAdow J, Rincon-Limas DE, Yin X, Tai W, Cheng G, Johnson A. Infection and chronic disease activate a systemic brain-muscle signaling axis. Sci Immunol 2024; 9:eadm7908. [PMID: 38996009 DOI: 10.1126/sciimmunol.adm7908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 04/18/2024] [Accepted: 06/18/2024] [Indexed: 07/14/2024]
Abstract
Infections and neurodegenerative diseases induce neuroinflammation, but affected individuals often show nonneural symptoms including muscle pain and muscle fatigue. The molecular pathways by which neuroinflammation causes pathologies outside the central nervous system (CNS) are poorly understood. We developed multiple models to investigate the impact of CNS stressors on motor function and found that Escherichia coli infections and SARS-CoV-2 protein expression caused reactive oxygen species (ROS) to accumulate in the brain. ROS induced expression of the cytokine Unpaired 3 (Upd3) in Drosophila and its ortholog, IL-6, in mice. CNS-derived Upd3/IL-6 activated the JAK-STAT pathway in skeletal muscle, which caused muscle mitochondrial dysfunction and impaired motor function. We observed similar phenotypes after expressing toxic amyloid-β (Aβ42) in the CNS. Infection and chronic disease therefore activate a systemic brain-muscle signaling axis in which CNS-derived cytokines bypass the connectome and directly regulate muscle physiology, highlighting IL-6 as a therapeutic target to treat disease-associated muscle dysfunction.
Collapse
Affiliation(s)
- Shuo Yang
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
- Department of Genetics and Genetics Engineering, School of Life Science, Fudan University, Shanghai 200438, China
| | - Meijie Tian
- Genetics Branch, Oncogenomics Section, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Yulong Dai
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen 518000, China
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Rong Wang
- Department of Genetics and Genetics Engineering, School of Life Science, Fudan University, Shanghai 200438, China
| | - Shigehiro Yamada
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Shengyong Feng
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Yunyun Wang
- Department of Forensic Medicine, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Deepak Chhangani
- Department of Neurology and McKnight Brain Institute, Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease, Genetics Institute, and Norman Fixel Institute for Neurological Diseases, University of Florida College of Medicine, Gainesville, FL 32611, USA
| | - Tiffany Ou
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Wenle Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Xuan Guo
- Life Science Institute, Jinzhou Medical University, Jinzhou 121001, China
| | - Jennifer McAdow
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Diego E Rincon-Limas
- Department of Neurology and McKnight Brain Institute, Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease, Genetics Institute, and Norman Fixel Institute for Neurological Diseases, University of Florida College of Medicine, Gainesville, FL 32611, USA
| | - Xin Yin
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Wanbo Tai
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen 518000, China
| | - Gong Cheng
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen 518000, China
- Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
- Southwest United Graduate School, Kunming 650092, China
| | - Aaron Johnson
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| |
Collapse
|
5
|
Islam MA, Sehar U, Sultana OF, Mukherjee U, Brownell M, Kshirsagar S, Reddy PH. SuperAgers and centenarians, dynamics of healthy ageing with cognitive resilience. Mech Ageing Dev 2024; 219:111936. [PMID: 38657874 DOI: 10.1016/j.mad.2024.111936] [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: 03/08/2024] [Revised: 04/08/2024] [Accepted: 04/16/2024] [Indexed: 04/26/2024]
Abstract
Graceful healthy ageing and extended longevity is the most desired goal for human race. The process of ageing is inevitable and has a profound impact on the gradual deterioration of our physiology and health since it triggers the onset of many chronic conditions like dementia, osteoporosis, diabetes, arthritis, cancer, and cardiovascular disease. However, some people who lived/live more than 100 years called 'Centenarians" and how do they achieve their extended lifespans are not completely understood. Studying these unknown factors of longevity is important not only to establish a longer human lifespan but also to manage and treat people with shortened lifespans suffering from age-related morbidities. Furthermore, older adults who maintain strong cognitive function are referred to as "SuperAgers" and may be resistant to risk factors linked to cognitive decline. Investigating the mechanisms underlying their cognitive resilience may contribute to the development of therapeutic strategies that support the preservation of cognitive function as people age. The key to a long, physically, and cognitively healthy life has been a mystery to scientists for ages. Developments in the medical sciences helps us to a better understanding of human physiological function and greater access to medical care has led us to an increase in life expectancy. Moreover, inheriting favorable genetic traits and adopting a healthy lifestyle play pivotal roles in promoting longer and healthier lives. Engaging in regular physical activity, maintaining a balanced diet, and avoiding harmful habits such as smoking contribute to overall well-being. The synergy between positive lifestyle choices, access to education, socio-economic factors, environmental determinants and genetic supremacy enhances the potential for a longer and healthier life. Our article aims to examine the factors associated with healthy ageing, particularly focusing on cognitive health in centenarians. We will also be discussing different aspects of ageing including genomic instability, metabolic burden, oxidative stress and inflammation, mitochondrial dysfunction, cellular senescence, immunosenescence, and sarcopenia.
Collapse
Affiliation(s)
- Md Ariful Islam
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Ujala Sehar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Omme Fatema Sultana
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Upasana Mukherjee
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Malcolm Brownell
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Sudhir Kshirsagar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Neurology, Departments of School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Nutritional Sciences Department, College of Human Sciences, Texas Tech University, 1301 Akron Ave, Lubbock, TX 79409, USA.
| |
Collapse
|
6
|
Raleigh SM, Orchard KJA. Sarcopenia as a Risk Factor for Alzheimer's Disease: Genetic and Epigenetic Perspectives. Genes (Basel) 2024; 15:561. [PMID: 38790190 PMCID: PMC11121242 DOI: 10.3390/genes15050561] [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: 03/28/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024] Open
Abstract
Sarcopenia, defined as the age-associated loss of muscle mass and increased fragility with age, is increasing worldwide. The condition often precedes the development of Alzheimer's disease, thereby decreasing the levels of mobility and physical activity in those affected. Indeed, the loss of muscle mass has, in some studies, been associated with an increased risk of Alzheimer's disease and other dementias. However, a detailed understanding of the interplay between both conditions is not available and needs to be thoroughly addressed. In the following review, we focus on several genes, specifically APOE, BDNF, ACE, FTO, and FNDC5, that have been associated with both conditions. We also discuss the epigenetic regulation of each of these genes along with non-coding RNAs (ncRNAs) that may have a role in the development of both the sarcopenic and Alzheimer's disease phenotypes. Finally, we assert that the application of systems biology will unravel the relationship between sarcopenia and Alzheimer's disease and believe that the prevention of muscle loss in older age will reduce the incidence of debilitating cognitive decline.
Collapse
Affiliation(s)
- Stuart M. Raleigh
- Centre for Health and Life Sciences, Coventry University, Coventry CV1 5FB, UK
| | - Kayleigh J. A. Orchard
- School of Life, Health and Chemical Sciences, Open University, Milton Keynes MK7 6AA, UK;
| |
Collapse
|
7
|
Paillard T, Blain H, Bernard PL. The impact of exercise on Alzheimer's disease progression. Expert Rev Neurother 2024; 24:333-342. [PMID: 38390841 DOI: 10.1080/14737175.2024.2319766] [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: 08/03/2023] [Accepted: 02/13/2024] [Indexed: 02/24/2024]
Abstract
INTRODUCTION The preventive effects of chronic physical exercise (CPE) on Alzheimer's disease (AD) are now admitted by the scientific community. Curative effects of CPE are more disputed, but they deserve to be investigated, since CPE is a natural non-pharmacological alternative for the treatment of AD. AREAS COVERED In this perspective, the authors discuss the impact of CPE on AD based on an exhaustive literature search using the electronic databases PubMed, ScienceDirect and Google Scholar. EXPERT OPINION Aerobic exercise alone is probably not the unique solution and needs to be complemented by other exercises (physical activities) to optimize the slowing down of AD. Anaerobic, muscle strength and power, balance/coordination and meditative exercises may also help to slow down the AD progression. However, the scientific evidence does not allow a precise description of the best training program for patients with AD. Influential environmental conditions (e.g. social relations, outdoor or indoor exercise) should also be studied to optimize training programs aimed at slowing down the AD progression.
Collapse
Affiliation(s)
- Thierry Paillard
- Movement, Balance, Performance, and Health Laboratory, Université de Pau & Pays de l'Adour, Tarbes, France
| | - Hubert Blain
- Pole de Gérontologie Antonin Balmes, CHU de Montpellier; EuroMov Digital Health in Motion, Univ Montpellier, IMT Mines Ales, Alès, France
| | - Pierre Louis Bernard
- UFR STAPS, EuroMov Digital Health in Motion, Université de Montpellier, IMT Mines Ales, Alès, France
| |
Collapse
|
8
|
Gao Y, Long Q, Yang H, Hu Y, Xu Y, Tang C, Gu C, Yong S. Transcriptomics and metabolomics study in mouse kidney of the molecular mechanism underlying energy metabolism response to hypoxic stress in highland areas. Exp Ther Med 2023; 26:533. [PMID: 37869643 PMCID: PMC10587886 DOI: 10.3892/etm.2023.12232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 08/25/2023] [Indexed: 10/24/2023] Open
Abstract
Exposure to hypoxia disrupts energy metabolism and induces inflammation. However, the pathways and mechanisms underlying energy metabolism disorders caused by hypoxic conditions remain unclear. In the present study, a hypoxic animal model was created and transcriptomic and non-targeted metabolomics techniques were applied to further investigate the pathways and mechanisms of hypoxia exposure that disrupt energy metabolism. Transcriptome results showed that 3,007 genes were significantly differentially expressed under hypoxic exposure, and Gene Ontology annotation analysis and Kyoto Encyclopaedia of Genes and Genomes (KEGG) enrichment analysis showed that the differentially expressed genes (DEGs) were mainly involved in energy metabolism and were significantly enriched in the tricarboxylic acid (TCA) cycle and oxidative phosphorylation (OXPHOS) pathway. The DEGs IDH3A, SUCLA2, and MDH2 in the TCA cycle and the DEGs NDUFA3, NDUFS7, UQCRC1, CYC1 and UQCRFS1 in the OXPHOS pathway were validated using mRNA and protein expression, and the results showed downregulation. The results of non-targeted metabolomics showed that 365 significant differential metabolites were identified under plateau hypoxia stress. KEGG enrichment analysis showed that the differential metabolites were mainly enriched in metabolic processes, such as energy, nucleotide and amino acid metabolism. Hypoxia exposure disrupted the TCA cycle and reduced the synthesis of amino acids and nucleotides by decreasing the concentration of cis-aconitate, α-ketoglutarate, NADH, NADPH and that of most amino acids, purines, and pyrimidines. Bioinformatics analysis was used to identify inflammatory genes related to hypoxia exposure and some of them were selected for verification. It was shown that the mRNA and protein expression levels of IL1B, IL12B, S100A8 and S100A9 in kidney tissues were upregulated under hypoxic exposure. The results suggest that hypoxia exposure inhibits the TCA cycle and the OXPHOS signalling pathway by inhibiting IDH3A, SUCLA2, MDH2, NDUFFA3, NDUFS7, UQCRC1, CYC1 and UQCRFS1, thereby suppressing energy metabolism, inducing amino acid and nucleotide deficiency and promoting inflammation, ultimately leading to kidney damage.
Collapse
Affiliation(s)
- Yujie Gao
- Department of Basic Medicine, School of Medicine, Qinghai University, Xining, Qinghai 810016, P.R. China
| | - Qifu Long
- Department of Basic Medicine, School of Medicine, Qinghai University, Xining, Qinghai 810016, P.R. China
| | - Hui Yang
- Department of Basic Medicine, School of Medicine, Qinghai University, Xining, Qinghai 810016, P.R. China
| | - Ying Hu
- Department of Basic Medicine, School of Medicine, Qinghai University, Xining, Qinghai 810016, P.R. China
| | - Yuzhen Xu
- Department of Basic Medicine, School of Medicine, Qinghai University, Xining, Qinghai 810016, P.R. China
| | - Chaoqun Tang
- Department of Basic Medicine, School of Medicine, Qinghai University, Xining, Qinghai 810016, P.R. China
| | - Cunlin Gu
- Department of Basic Medicine, School of Medicine, Qinghai University, Xining, Qinghai 810016, P.R. China
| | - Sheng Yong
- Department of Basic Medicine, School of Medicine, Qinghai University, Xining, Qinghai 810016, P.R. China
| |
Collapse
|
9
|
Shen ZL, Chen WH, Liu Z, Yu DY, Chen WZ, Zang WF, Zhang P, Yan XL, Yu Z. A novel insight into the key gene signature associated with the immune landscape in the progression of sarcopenia. Exp Gerontol 2023; 179:112244. [PMID: 37343810 DOI: 10.1016/j.exger.2023.112244] [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: 04/06/2023] [Revised: 06/04/2023] [Accepted: 06/18/2023] [Indexed: 06/23/2023]
Abstract
Sarcopenia is an age-related skeletal muscle disorder that causes falls, disability and death in the elderly, but its exact mechanism remains unknown. In this study, we merged three GEO datasets into the expression profiles of 118 samples and screened 22 differentially expressed genes (DEGs) as candidate genes. Pathway analysis demonstrated that the functional enrichment of DEGs is mainly in the cellular response to insulin stimulus, PPAR signaling pathway and other metabolism-related pathways. Then, we identified six key genes by machine learning, which were confirmed to be closely associated with sarcopenia by bioinformatics analysis. It was experimentally verified that SCD1 exhibits the most substantial alterations in the progression of sarcopenia with disturbed lipid metabolism and myosteatosis. In addition, the immune microenvironment of sarcopenia was found to be affected by these key genes, with Th17 cells down-regulated and NK cells up-regulated. Sarcopenic patients consequently presented a more significant systemic inflammatory state with higher CAR (p = 0.028) and PAR (p = 0.018). For the first time, we identified key genes in sarcopenia with high-throughput data and demonstrated that key genes can regulate the progression of sarcopenia by affecting the immune microenvironment. Among them, SCD1 may influence lipid metabolism and myosteatosis process. Screening of key genes and analyzing of immune microenvironment provide a more accurate target for treating sarcopenia.
Collapse
Affiliation(s)
- Zi-Le Shen
- Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Wen-Hao Chen
- Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China; Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Zhang Liu
- Department of Cardio-Thoracic Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Ding-Ye Yu
- Department of General Surgery, Huadong Hospital, Fudan University, Shanghai 200040, China
| | - Wei-Zhe Chen
- Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Wang-Fu Zang
- Department of Cardio-Thoracic Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Peng Zhang
- Department of Cardio-Thoracic Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China.
| | - Xia-Lin Yan
- Department of Colorectal Anal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| | - Zhen Yu
- Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China; Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| |
Collapse
|
10
|
Yu Z, Li M, Peng W. Exploring biomarkers of premature ovarian insufficiency based on oxford nanopore transcriptional profile and machine learning. Sci Rep 2023; 13:11498. [PMID: 37460774 PMCID: PMC10352282 DOI: 10.1038/s41598-023-38754-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 07/14/2023] [Indexed: 07/20/2023] Open
Abstract
Premature ovarian insufficiency (POI) is a reproductive endocrine disorder characterized by infertility and perimenopausal syndrome, with a highly heterogeneous genetic etiology and its mechanism is not fully understood. Therefore, we utilized Oxford Nanopore Technology (ONT) for the first time to characterize the full-length transcript profile, and revealed biomarkers, pathway and molecular mechanisms for POI by bioinformatics analysis and machine learning. Ultimately, we identified 272 differentially expressed genes, 858 core genes, and 25 hub genes by analysis of differential expression, gene set enrichment, and protein-protein interactions. Seven candidate genes were identified based on the intersection features of the random forest and Boruta algorithm. qRT-PCR results indicated that COX5A, UQCRFS1, LCK, RPS2 and EIF5A exhibited consistent expression trends with sequencing data and have potential as biomarkers. Additionally, GSEA analysis revealed that the pathophysiology of POI is closely associated with inhibition of the PI3K-AKT pathway, oxidative phosphorylation and DNA damage repair, as well as activation of inflammatory and apoptotic pathways. Furthermore, we emphasize that downregulation of respiratory chain enzyme complex subunits and inhibition of oxidative phosphorylation pathways play crucial roles in the pathophysiology of POI. In conclusion, our utilization of long-read sequencing has refined the annotation information within the POI transcriptional profile. This valuable data provides novel insights for further exploration into molecular regulatory networks and potential biomarkers associated with POI.
Collapse
Affiliation(s)
- Zhaoyang Yu
- The First Affiliated Clinical College of Guangxi Medical University, Nanning, China
| | - Mujun Li
- Reproductive Medicine Research Center, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.
| | - Weilong Peng
- School of Computer Science and Cyber Engineering, Guangzhou University, Guangzhou, China.
| |
Collapse
|
11
|
Wu Z, Liu P, Huang B, Deng S, Song Z, Huang X, Yang J, Cheng S. A novel Alzheimer's disease prognostic signature: identification and analysis of glutamine metabolism genes in immunogenicity and immunotherapy efficacy. Sci Rep 2023; 13:6895. [PMID: 37106067 PMCID: PMC10140060 DOI: 10.1038/s41598-023-33277-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Alzheimer's disease (AD) is characterized as a distinct onset and progression of cognitive and functional decline associated with age, as well as a specific neuropathology. It has been discovered that glutamine (Gln) metabolism plays a crucial role in cancer. However, a full investigation of its role in Alzheimer's disease is still missing. This study intended to find and confirm potential Gln-related genes associated with AD using bioinformatics analysis. The discovery of GlnMgs was made possible by the intersection of the WGCNA test and 26 Gln-metabolism genes (GlnMgs). GlnMgs' putative biological functions and pathways were identified using GSVA. The LASSO method was then used to identify the hub genes as well as the diagnostic efficiency of the four GlnMgs in identifying AD. The association between hub GlnMgs and clinical characteristics was also studied. Finally, the GSE63060 was utilized to confirm the levels of expression of the four GlnMgs. Four GlnMgs were discovered (ATP5H, NDUFAB1, PFN2, and SPHKAP). For biological function analysis, cell fate specification, atrioventricular canal development, and neuron fate specification were emphasized. The diagnostic ability of the four GlnMgs in differentiating AD exhibited a good value. This study discovered four GlnMgs that are linked to AD. They shed light on potential new biomarkers for AD and tracking its progression.
Collapse
Affiliation(s)
- Zixuan Wu
- Hunan University of Chinese Medicine, Changsha, 410128, China
| | - Ping Liu
- Hunan University of Chinese Medicine, Changsha, 410128, China
| | - Baisheng Huang
- Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Sisi Deng
- Hunan University of Chinese Medicine, Changsha, 410128, China
| | - Zhenyan Song
- Hunan University of Chinese Medicine, Changsha, 410128, China
| | - Xindi Huang
- Hunan University of Chinese Medicine, Changsha, 410128, China
| | - Jing Yang
- Hunan University of Chinese Medicine, Changsha, 410128, China.
| | - Shaowu Cheng
- Hunan University of Chinese Medicine, Changsha, 410128, China.
| |
Collapse
|
12
|
Parnell LD, Magadmi R, Zwanger S, Shukitt-Hale B, Lai CQ, Ordovás JM. Dietary Responses of Dementia-Related Genes Encoding Metabolic Enzymes. Nutrients 2023; 15:644. [PMID: 36771351 PMCID: PMC9921944 DOI: 10.3390/nu15030644] [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] [Received: 09/29/2022] [Revised: 01/19/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023] Open
Abstract
The age-related loss of the cognitive function is a growing concern for global populations. Many factors that determine cognitive resilience or dementia also have metabolic functions. However, this duality is not universally appreciated when the action of that factor occurs in tissues external to the brain. Thus, we examined a set of genes involved in dementia, i.e., those related to vascular dementia, Alzheimer's disease, Parkinson's disease, and the human metabolism for activity in 12 metabolically active tissues. Mining the Genotype-Tissue Expression (GTEx) data showed that most of these metabolism-dementia (MD) genes (62 of 93, 67%) exhibit a higher median expression in any of the metabolically active tissues than in the brain. After identifying that several MD genes served as blood-based biomarkers of longevity in other studies, we examined the impact of the intake of food, nutrients, and other dietary factors on the expression of MD genes in whole blood in the Framingham Offspring Study (n = 2134). We observed positive correlations between flavonoids and HMOX1, taurine and UQCRC1, broccoli and SLC10A2, and myricetin and SLC9A8 (p < 2.09 × 10-4). In contrast, dairy protein, palmitic acid, and pie were negatively correlated, respectively, with the expression of IGF1R, CSF1R, and SLC9A8, among others (p < 2.92 × 10-4). The results of this investigation underscore the potential contributions of metabolic enzyme activity in non-brain tissues to the risk of dementia. Specific epidemiological or intervention studies could be designed using specific foods and nutrients or even dietary patterns focused on these foods and nutrients that influence the expression of some MD genes to verify the findings presented here.
Collapse
Affiliation(s)
- Laurence D Parnell
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Agricultural Research Service, US Department of Agriculture, Boston, MA 02111, USA
| | - Rozana Magadmi
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA 02111, USA
| | | | - Barbara Shukitt-Hale
- Neuroscience and Aging Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Agricultural Research Service, US Department of Agriculture, Boston, MA 02111, USA
| | - Chao-Qiang Lai
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Agricultural Research Service, US Department of Agriculture, Boston, MA 02111, USA
| | - José M Ordovás
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA 02111, USA
| |
Collapse
|
13
|
Alizadeh R, Salehi O, Rezaeinezhad N, Hosseini SA. The effect of high intensity interval training with genistein supplementation on mitochondrial function in the heart tissue of elderly rats. Exp Gerontol 2023; 171:112039. [PMID: 36442700 DOI: 10.1016/j.exger.2022.112039] [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: 04/09/2022] [Revised: 11/08/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022]
Abstract
INTRODUCTION For the most part, heart disease increases with age; on the other hand, although the role of exercise and antioxidants in the health of the elderly has been reported, the simultaneous effect of these two interventions is a new research topic. Thus, the aim of this study was to evaluate the effect of eight weeks of high intensity interval training (HIIT) and genistein (G) supplementation on oxidative stress, apoptosis and mitochondrial biogenesis in the heart tissue of elderly rats. METHODS In this experimental study, 40 elderly female Sprague-Dawley rats aged 20 ± 2 months and weighing 250 ± 30 g were randomly divided into five groups of eight animals, including: (1) control (C), (2) sham (Sh), (3) HIIT, (4) HIIT + G and (5) G. Also, to evaluate the effect of time passage on the variables, 8 healthy young rats were included in the healthy young control group. HIIT protocol was performed for eight weeks, three sessions with an intensity of 95-90 % VO2max at high intensity intervals and 45-45 % VO2max at low intensity intervals. Ge was received daily at a dose of 60 mg/kg peritoneally. Data analysis was performed using one-way ANOVA with Tukey's post hoc test (P ≤ 0.05). RESULTS In the HIIT and HIIT + G groups, levels of Bax, Bax/Bcl-2 ratio, MDA, PAB, GSSG were lower and levels of PGC-1α, TFAM, GSH, GSH/GSSG ratio and NDUFS7 were higher than the control and sham groups (P ≤ 0.05). In the HIIT + G group, levels of Bcl-2 were significantly higher than the control group (P ≤ 0.05). In the HIIT + G group, levels of Bax, GSSG, Bax/Bcl-2 ratio, and PAB were lower, and levels of GSH/GSSG ratio, Bcl-2, PGC-1α, TFAM and NDUFS7 were higher than the G consumption group (P ≤ 0.05). In the HIIT + G group, levels of PGC-1α and TFAM were significantly higher and levels of MDA and PAB were lower than the HIIT group (P ≤ 0.05). CONCLUSION Both HIIT and G consumption seem to have beneficial effects on reducing oxidative stress; in addition, the interaction of these two variables on the improvement of apoptosis and mitochondrial biogenesis is more favorable than the effect of either one alone. However, more studies are needed on different pathways of apoptosis following G administration.
Collapse
Affiliation(s)
- Rostam Alizadeh
- Department of Sports Science, School of Literature and Humanities, Ilam University, Ilam, Iran.
| | - Omidreza Salehi
- Department of Physical Education and Sport Sciences, University of Kurdistan, Sanandaj, Iran
| | | | - Seyed Ali Hosseini
- Department of Sport Physiology, Marvdasht Branch, Islamic Azad University, Marvdasht, Iran.
| |
Collapse
|
14
|
Radiotherapy Side Effects: Comprehensive Proteomic Study Unraveled Neural Stem Cell Degenerative Differentiation upon Ionizing Radiation. Biomolecules 2022; 12:biom12121759. [PMID: 36551187 PMCID: PMC9775306 DOI: 10.3390/biom12121759] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 11/11/2022] [Accepted: 11/15/2022] [Indexed: 11/29/2022] Open
Abstract
Cranial radiation therapy is one of the most effective treatments for childhood brain cancers. Despite the ameliorated survival rate of juvenile patients, radiation exposure-induced brain neurogenic region injury could markedly impair patients' cognitive functions and even their quality of life. Determining the mechanism underlying neural stem cells (NSCs) response to irradiation stress is a crucial therapeutic strategy for cognitive impairment. The present study demonstrated that X-ray irradiation arrested NSCs' cell cycle and impacted cell differentiation. To further characterize irradiation-induced molecular alterations in NSCs, two-dimensional high-resolution mass spectrometry-based quantitative proteomics analyses were conducted to explore the mechanism underlying ionizing radiation's influence on stem cell differentiation. We observed that ionizing radiation suppressed intracellular protein transport, neuron projection development, etc., particularly in differentiated cells. Redox proteomics was performed for the quantification of cysteine thiol modifications in order to profile the oxidation-reduction status of proteins in stem cells that underwent ionizing radiation treatment. Via conjoint screening of protein expression abundance and redox status datasets, several significantly expressed and oxidized proteins were identified in differentiating NSCs subjected to X-ray irradiation. Among these proteins, succinate dehydrogenase [ubiquinone] flavoprotein subunit, mitochondrial (sdha) and the acyl carrier protein, mitochondrial (Ndufab1) were highly related to neurodegenerative diseases such as Parkinson's disease, Alzheimer's disease, and Huntington's disease, illustrating the dual-character of NSCs in cell differentiation: following exposure to ionizing radiation, the normal differentiation of NSCs was compromised, and the upregulated oxidized proteins implied a degenerative differentiation trajectory. These findings could be integrated into research on neurodegenerative diseases and future preventive strategies.
Collapse
|