1
|
Wu X, Chen M, Liu K, Wu Y, Feng Y, Fu S, Xu H, Zhao Y, Lin F, Lin L, Ye S, Lin J, Xiao T, Li W, Lou M, Lv H, Qiu Y, Yu R, Chen W, Li M, Feng X, Luo Z, Guo L, Ke H, Zhao L. Molecular classification of geriatric breast cancer displays distinct senescent subgroups of prognostic significance. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102309. [PMID: 39296329 PMCID: PMC11408383 DOI: 10.1016/j.omtn.2024.102309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 08/12/2024] [Indexed: 09/21/2024]
Abstract
Breast cancer in the elderly presents distinct biological characteristics and clinical treatment responses compared with cancer in younger patients. Comprehensive Geriatric Assessment is recommended for evaluating treatment efficacy in elderly cancer patients based on physiological classification. However, research on molecular classification in older cancer patients remains insufficient. In this study, we identified two subgroups with distinct senescent clusters among geriatric breast cancer patients through multi-omics analysis. Using various machine learning algorithms, we developed a comprehensive scoring model called "Sene_Signature," which more accurately distinguished elderly breast cancer patients compared with existing methods and better predicted their prognosis. The Sene_Signature was correlated with tumor immune cell infiltration, as supported by single-cell transcriptomics, RNA sequencing, and pathological data. Furthermore, we observed increased drug responsiveness in patients with a high Sene_Signature to treatments targeting the epidermal growth factor receptor and cell-cycle pathways. We also established a user-friendly web platform to assist investigators in assessing Sene_Signature scores and predicting treatment responses for elderly breast cancer patients. In conclusion, we developed a novel model for evaluating prognosis and therapeutic responses, providing a potential molecular classification that assists in the pre-treatment assessment of geriatric breast cancer.
Collapse
Affiliation(s)
- Xia Wu
- Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China
- Ningbo Clinical Pathology Diagnosis Center, Ningbo, Zhejiang 315021, China
| | - Mengxin Chen
- Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China
| | - Kang Liu
- Ganzhou People's Hospital, Ganzhou 341000, China
| | - Yixin Wu
- Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China
| | - Yun Feng
- Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China
| | - Shiting Fu
- Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China
| | - Huaimeng Xu
- Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China
| | - Yongqi Zhao
- Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China
| | - Feilong Lin
- Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China
| | - Liang Lin
- Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China
| | - Shihui Ye
- Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China
| | - Junqiang Lin
- Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China
| | - Taiping Xiao
- Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China
| | - Wenhao Li
- Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China
| | - Meng Lou
- Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China
| | - Hongyu Lv
- Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China
| | - Ye Qiu
- Huankui Academy, Nanchang University, Nanchang, Jiangxi 330031, China
| | - Ruifan Yu
- Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China
| | - Wenyan Chen
- Department of Medical Oncology, Nanchang People's Hospital, Nanchang 330008, China
| | - Mengyuan Li
- Department of Gynaecology and Obstetrics, Chongqing General Hospital, Chongqing 401147, China
| | - Xu Feng
- Xianlin High School, Weinan 714000, China
| | | | - Lu Guo
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310000, China
| | - Hao Ke
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, China
- School of Life Science, Nanchang University, Nanchang 330031, China
| | - Limin Zhao
- Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China
| |
Collapse
|
2
|
Vazquez JM, Lauterbur ME, Mottaghinia S, Bucci M, Fraser D, Gray-Sandoval G, Gaucherand L, Haidar ZR, Han M, Kohler W, Lama TM, Le Corf A, Loyer C, Maesen S, McMillan D, Li S, Lo J, Rey C, Capel SLR, Singer M, Slocum K, Thomas W, Tyburec JD, Villa S, Miller R, Buchalski M, Vazquez-Medina JP, Pfeffer S, Etienne L, Enard D, Sudmant PH. Extensive longevity and DNA virus-driven adaptation in nearctic Myotis bats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.10.617725. [PMID: 39416019 PMCID: PMC11482938 DOI: 10.1101/2024.10.10.617725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
The genus Myotis is one of the largest clades of bats, and exhibits some of the most extreme variation in lifespans among mammals alongside unique adaptations to viral tolerance and immune defense. To study the evolution of longevity-associated traits and infectious disease, we generated near-complete genome assemblies and cell lines for 8 closely related species of Myotis. Using genome-wide screens of positive selection, analyses of structural variation, and functional experiments in primary cell lines, we identify new patterns of adaptation contributing to longevity, cancer resistance, and viral interactions in bats. We find that Myotis bats have some of the most significant variation in cancer risk across mammals and demonstrate a unique DNA damage response in primary cells of the long-lived M. lucifugus. We also find evidence of abundant adaptation in response to DNA viruses - but not RNA viruses - in Myotis and other bats in sharp contrast with other mammals, potentially contributing to the role of bats as reservoirs of zoonoses. Together, our results demonstrate how genomics and primary cells derived from diverse taxa uncover the molecular bases of extreme adaptations in non-model organisms.
Collapse
Affiliation(s)
- Juan M Vazquez
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA USA
- These authors contributed equally
| | - M. Elise Lauterbur
- Department of Ecology and Evolutionary Biology, University of Arizona, Tucson, AZ USA
- Current affiliation: Department of Biology, University of Vermont, Burlington, VT USA
- These authors contributed equally
| | - Saba Mottaghinia
- Centre International de Recherche en Infectiologie (CIRI), Inserm U1111, UCBL1, CNRS UMR5308, Ecole Normale Supérieure ENS de Lyon, Université de Lyon, Lyon, France
| | - Melanie Bucci
- Department of Ecology and Evolutionary Biology, University of Arizona, Tucson, AZ USA
| | - Devaughn Fraser
- Wildlife Genetics Research Unit, Wildlife Health Laboratory, California Department of Fish and Wildlife, Sacramento, CA, United States
- Current affiliation: Wildlife Diversity Program, Wildlife Division, Connecticut Department of Energy and Environmental Protection, Burlington, CT, United States
| | | | - Léa Gaucherand
- Université de Strasbourg, Architecture et Réactivité de l’ARN, Institut de Biologie Moléculaire et Cellulaire du CNRS, Strasbourg, France
| | - Zeinab R Haidar
- Department of Biology, California State Polytechnic University, Humboldt, Arcata, CA USA
- Current affiliation: Western EcoSystems Technology Inc, Cheyenne, WY USA
| | - Melissa Han
- Department of Pathology and Clinical Laboratories, University of Michigan, Ann Arbor, MI USA
| | - William Kohler
- Department of Pathology and Clinical Laboratories, University of Michigan, Ann Arbor, MI USA
| | - Tanya M. Lama
- Department of Biological Sciences, Smith College, Northampton, MA USA
| | - Amandine Le Corf
- Centre International de Recherche en Infectiologie (CIRI), Inserm U1111, UCBL1, CNRS UMR5308, Ecole Normale Supérieure ENS de Lyon, Université de Lyon, Lyon, France
| | - Clara Loyer
- Centre International de Recherche en Infectiologie (CIRI), Inserm U1111, UCBL1, CNRS UMR5308, Ecole Normale Supérieure ENS de Lyon, Université de Lyon, Lyon, France
| | - Sarah Maesen
- Centre International de Recherche en Infectiologie (CIRI), Inserm U1111, UCBL1, CNRS UMR5308, Ecole Normale Supérieure ENS de Lyon, Université de Lyon, Lyon, France
| | - Dakota McMillan
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA USA
- Department of Science and Biotechnology, Berkeley City College, Berkeley, CA USA
| | - Stacy Li
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA USA
- Center for Computational Biology, University of California, Berkeley, Berkeley, CA USA
| | - Johnathan Lo
- Center for Computational Biology, University of California, Berkeley, Berkeley, CA USA
| | - Carine Rey
- Centre International de Recherche en Infectiologie (CIRI), Inserm U1111, UCBL1, CNRS UMR5308, Ecole Normale Supérieure ENS de Lyon, Université de Lyon, Lyon, France
| | - Samantha LR Capel
- Current affiliation: Wildlife Diversity Program, Wildlife Division, Connecticut Department of Energy and Environmental Protection, Burlington, CT, United States
| | - Michael Singer
- Department of Molecular and Cellular Biology, University of California, Berkeley, Berkeley, CA USA
| | | | - William Thomas
- Department of Ecology and Evolution, Stony Brook University, Stony Brook NY USA
| | | | - Sarah Villa
- Department of Molecular and Cellular Biology, University of California, Berkeley, Berkeley, CA USA
| | - Richard Miller
- Department of Pathology and Clinical Laboratories, University of Michigan, Ann Arbor, MI USA
| | - Michael Buchalski
- Wildlife Genetics Research Unit, Wildlife Health Laboratory, California Department of Fish and Wildlife, Sacramento, CA, United States
| | | | - Sébastien Pfeffer
- Université de Strasbourg, Architecture et Réactivité de l’ARN, Institut de Biologie Moléculaire et Cellulaire du CNRS, Strasbourg, France
| | - Lucie Etienne
- Centre International de Recherche en Infectiologie (CIRI), Inserm U1111, UCBL1, CNRS UMR5308, Ecole Normale Supérieure ENS de Lyon, Université de Lyon, Lyon, France
- Senior author
| | - David Enard
- Department of Ecology and Evolutionary Biology, University of Arizona, Tucson, AZ USA
- Senior author
- These authors contributed equally
| | - Peter H Sudmant
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA USA
- Center for Computational Biology, University of California, Berkeley, Berkeley, CA USA
- Senior author
- These authors contributed equally
- Lead contact
| |
Collapse
|
3
|
Cooper LN, Ansari MY, Capshaw G, Galazyuk A, Lauer AM, Moss CF, Sears KE, Stewart M, Teeling EC, Wilkinson GS, Wilson RC, Zwaka TP, Orman R. Bats as instructive animal models for studying longevity and aging. Ann N Y Acad Sci 2024; 1541:10-23. [PMID: 39365995 PMCID: PMC11580778 DOI: 10.1111/nyas.15233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/06/2024]
Abstract
Bats (order Chiroptera) are emerging as instructive animal models for aging studies. Unlike some common laboratory species, they meet a central criterion for aging studies: they live for a long time in the wild or in captivity, for 20, 30, and even >40 years. Healthy aging (i.e., healthspan) in bats has drawn attention to their potential to improve the lives of aging humans due to bat imperviousness to viral infections, apparent low rate of tumorigenesis, and unique ability to repair DNA. At the same time, bat longevity also permits the accumulation of age-associated systemic pathologies that can be examined in detail and manipulated, especially in captive animals. Research has uncovered additional and critical advantages of bats. In multiple ways, bats are better analogs to humans than are rodents. In this review, we highlight eight diverse areas of bat research with relevance to aging: genome sequencing, telomeres, and DNA repair; immunity and inflammation; hearing; menstruation and menopause; skeletal system and fragility; neurobiology and neurodegeneration; stem cells; and senescence and mortality. These examples demonstrate the broad relevance of the bat as an animal model and point to directions that are particularly important for human aging studies.
Collapse
Affiliation(s)
- Lisa Noelle Cooper
- Department of Anatomy and NeurobiologyNortheast Ohio Medical UniversityRootstownOhioUSA
| | - Mohammad Y. Ansari
- Department of Anatomy and NeurobiologyNortheast Ohio Medical UniversityRootstownOhioUSA
| | - Grace Capshaw
- Department of Psychological and Brain SciencesJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Alex Galazyuk
- Department of Anatomy and NeurobiologyNortheast Ohio Medical UniversityRootstownOhioUSA
| | - Amanda M. Lauer
- Department of Otolaryngology – HNSJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Cynthia F. Moss
- Department of Psychological and Brain SciencesJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Karen E. Sears
- Department of Ecology and Evolutionary Biology, Department of Molecular, Cellular, and Developmental BiologyUniversity of California Los AngelesLos AngelesCaliforniaUSA
| | - Mark Stewart
- Department of Physiology & PharmacologySUNY Downstate Health Sciences UniversityBrooklynNew YorkUSA
| | - Emma C. Teeling
- School of Biology and Environmental Science, Science Centre EastUniversity College DublinDublinIreland
| | - Gerald S. Wilkinson
- Department of BiologyUniversity of Maryland at College ParkCollege ParkMarylandUSA
| | | | - Thomas P. Zwaka
- Black Family Stem Cell Institute, Huffington Center for Cell‐based Research in Parkinson's Disease, Department of Cell, Developmental and Regenerative BiologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Rena Orman
- Department of Physiology & PharmacologySUNY Downstate Health Sciences UniversityBrooklynNew YorkUSA
| |
Collapse
|
4
|
Danis T, Rokas A. The evolution of gestation length in eutherian mammals. Proc Biol Sci 2024; 291:20241412. [PMID: 39471860 PMCID: PMC11521618 DOI: 10.1098/rspb.2024.1412] [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: 01/22/2024] [Revised: 08/16/2024] [Accepted: 09/25/2024] [Indexed: 11/01/2024] Open
Abstract
Eutherian mammals exhibit considerable variation in their gestation lengths, which has traditionally been linked to variation in other traits, including body mass and lifespan. To understand how gestation length variation, including its association with body mass and lifespan variation, changed over mammalian evolution, we conducted phylogeny-informed analyses of 845 representative extant species. We found that gestation length substantially differed in both whether and how strongly it was associated with body mass and lifespan across mammals. For example, gestation length was not associated with lifespan or body mass in Chiroptera and Cetacea but was strongly associated only with body mass in Carnivora. We also identified 52 evolutionary shifts in gestation length variation across the mammal phylogeny and 14 shifts when we jointly considered variation of all three traits; six shifts were shared. Notably, two of these shifts, both positive, occurred at the roots of Cetacea and Pinnipedia, respectively, coinciding with the transition of these clades to the marine environment, whereas a negative shift occurred at the root of Chiroptera, coinciding with the evolution of flight in this clade. These results suggest that the relationship between gestation length and the two other traits has varied substantially across mammalian phylogeny.
Collapse
Affiliation(s)
- Thodoris Danis
- Department of Biological Sciences, Vanderbilt University, Nashville, TN37235, USA
- Evolutionary Studies Initiative, Vanderbilt University, Nashville, TN37235, USA
| | - Antonis Rokas
- Department of Biological Sciences, Vanderbilt University, Nashville, TN37235, USA
- Evolutionary Studies Initiative, Vanderbilt University, Nashville, TN37235, USA
| |
Collapse
|
5
|
Berv JS, Singhal S, Field DJ, Walker-Hale N, McHugh SW, Shipley JR, Miller ET, Kimball RT, Braun EL, Dornburg A, Parins-Fukuchi CT, Prum RO, Winger BM, Friedman M, Smith SA. Genome and life-history evolution link bird diversification to the end-Cretaceous mass extinction. SCIENCE ADVANCES 2024; 10:eadp0114. [PMID: 39083615 PMCID: PMC11290531 DOI: 10.1126/sciadv.adp0114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 06/28/2024] [Indexed: 08/02/2024]
Abstract
Complex patterns of genome evolution associated with the end-Cretaceous [Cretaceous-Paleogene (K-Pg)] mass extinction limit our understanding of the early evolutionary history of modern birds. Here, we analyzed patterns of avian molecular evolution and identified distinct macroevolutionary regimes across exons, introns, untranslated regions, and mitochondrial genomes. Bird clades originating near the K-Pg boundary exhibited numerous shifts in the mode of molecular evolution, suggesting a burst of genomic heterogeneity at this point in Earth's history. These inferred shifts in substitution patterns were closely related to evolutionary shifts in developmental mode, adult body mass, and patterns of metabolic scaling. Our results suggest that the end-Cretaceous mass extinction triggered integrated patterns of evolution across avian genomes, physiology, and life history near the dawn of the modern bird radiation.
Collapse
Affiliation(s)
- Jacob S. Berv
- Department of Ecology and Evolutionary Biology, University of Michigan, 1105 North University Avenue, Biological Sciences Building, University of Michigan, Ann Arbor, MI 48109, USA
- Museum of Paleontology, University of Michigan, 1105 North University Avenue, Biological Sciences Building, University of Michigan, Ann Arbor, MI 48109, USA
- Museum of Zoology, University of Michigan, 1105 North University Avenue, Biological Sciences Building, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sonal Singhal
- Department of Biology, California State University, Dominguez Hills, Carson, CA 90747, USA
| | - Daniel J. Field
- Department of Earth Sciences, University of Cambridge, Downing Street, Cambridge CB2 3EQ, UK
- Museum of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK
| | - Nathanael Walker-Hale
- Department of Plant Sciences, University of Cambridge, Downing Street, Cambridge CB2 3EA, UK
| | - Sean W. McHugh
- Department of Evolution, Ecology, and Population Biology, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - J. Ryan Shipley
- Department of Forest Dynamics, Swiss Federal Institute for Forest, Snow, and Landscape Research WSL, Zürcherstrasse 111 8903, Birmensdorf, Switzerland
| | - Eliot T. Miller
- Center for Avian Population Studies, Cornell Lab of Ornithology, Cornell University, Ithaca, NY 14850, USA
| | - Rebecca T. Kimball
- Department of Biology, University of Florida, Gainesville, FL 32611, USA
| | - Edward L. Braun
- Department of Biology, University of Florida, Gainesville, FL 32611, USA
| | - Alex Dornburg
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - C. Tomomi Parins-Fukuchi
- Department of Ecology and Evolutionary Biology, University of Toronto, Toronto, Ontario M5S 3B2, Canada
| | - Richard O. Prum
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT 06520, USA
- Peabody Museum of Natural History, Yale University, New Haven, CT 06520, USA
| | - Benjamin M. Winger
- Department of Ecology and Evolutionary Biology, University of Michigan, 1105 North University Avenue, Biological Sciences Building, University of Michigan, Ann Arbor, MI 48109, USA
- Museum of Zoology, University of Michigan, 1105 North University Avenue, Biological Sciences Building, University of Michigan, Ann Arbor, MI 48109, USA
| | - Matt Friedman
- Museum of Paleontology, University of Michigan, 1105 North University Avenue, Biological Sciences Building, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Earth and Environmental Sciences, University of Michigan, 1100 North University Avenue, University of Michigan, Ann Arbor, MI 48109, USA
| | - Stephen A. Smith
- Department of Ecology and Evolutionary Biology, University of Michigan, 1105 North University Avenue, Biological Sciences Building, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
6
|
Lukacsovich D, O’Shea D, Huang H, Zhang W, Young J, Chen XS, Dietrich ST, Kunkle B, Martin E, Wang L. MIAMI-AD (Methylation in Aging and Methylation in AD): an integrative knowledgebase that facilitates explorations of DNA methylation across sex, aging, and Alzheimer's disease. Database (Oxford) 2024; 2024:baae061. [PMID: 39028752 PMCID: PMC11259044 DOI: 10.1093/database/baae061] [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: 01/19/2024] [Revised: 05/08/2024] [Accepted: 07/03/2024] [Indexed: 07/21/2024]
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disorder with a significant impact on aging populations. DNA methylation (DNAm) alterations have been implicated in both the aging processes and the development of AD. Given that AD affects more women than men, it is also important to explore DNAm changes that occur specifically in each sex. We created MIAMI-AD, a comprehensive knowledgebase containing manually curated summary statistics from 98 published tables in 38 studies, all of which included at least 100 participants. MIAMI-AD enables easy browsing, querying, and downloading DNAm associations at multiple levels-at individual CpG, gene, genomic regions, or genome-wide, in one or multiple studies. Moreover, it also offers tools to perform integrative analyses, such as comparing DNAm associations across different phenotypes or tissues, as well as interactive visualizations. Using several use case examples, we demonstrated that MIAMI-AD facilitates our understanding of age-associated CpGs in AD and the sex-specific roles of DNAm in AD. This open-access resource is freely available to the research community, and all the underlying data can be downloaded. MIAMI-AD facilitates integrative explorations to better understand the interplay between DNAm across aging, sex, and AD. Database URL: https://miami-ad.org/.
Collapse
Affiliation(s)
- David Lukacsovich
- Division of Biostatistics, Department of Public Health Sciences, University of Miami, Miller School of Medicine, 1120 NW 14th Street, Miami, FL 33136, USA
| | - Deirdre O’Shea
- Department of Neurology, Comprehensive Center for Brain Health, University of Miami, Miller School of Medicine, 7700 W Camino Real, Boca Raton, FL 33433, USA
| | - Hanchen Huang
- Division of Biostatistics, Department of Public Health Sciences, University of Miami, Miller School of Medicine, 1120 NW 14th Street, Miami, FL 33136, USA
| | - Wei Zhang
- Division of Biostatistics, Department of Public Health Sciences, University of Miami, Miller School of Medicine, 1120 NW 14th Street, Miami, FL 33136, USA
| | - Juan Young
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, 1501 NW 10th Ave, Miami, FL 33136, USA
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, 1501 NW 10th Ave, Miami, FL 33136, USA
| | - X Steven Chen
- Division of Biostatistics, Department of Public Health Sciences, University of Miami, Miller School of Medicine, 1120 NW 14th Street, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, 1475 NW 12th Ave, Miami, FL 33136, USA
| | - Sven-Thorsten Dietrich
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, 1501 NW 10th Ave, Miami, FL 33136, USA
| | - Brian Kunkle
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, 1501 NW 10th Ave, Miami, FL 33136, USA
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, 1501 NW 10th Ave, Miami, FL 33136, USA
| | - Eden Martin
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, 1501 NW 10th Ave, Miami, FL 33136, USA
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, 1501 NW 10th Ave, Miami, FL 33136, USA
| | - Lily Wang
- Division of Biostatistics, Department of Public Health Sciences, University of Miami, Miller School of Medicine, 1120 NW 14th Street, Miami, FL 33136, USA
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, 1501 NW 10th Ave, Miami, FL 33136, USA
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, 1501 NW 10th Ave, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, 1475 NW 12th Ave, Miami, FL 33136, USA
| |
Collapse
|
7
|
Pérez RF, Tezanos P, Peñarroya A, González-Ramón A, Urdinguio RG, Gancedo-Verdejo J, Tejedor JR, Santamarina-Ojeda P, Alba-Linares JJ, Sainz-Ledo L, Roberti A, López V, Mangas C, Moro M, Cintado Reyes E, Muela Martínez P, Rodríguez-Santamaría M, Ortea I, Iglesias-Rey R, Castilla-Silgado J, Tomás-Zapico C, Iglesias-Gutiérrez E, Fernández-García B, Sanchez-Mut JV, Trejo JL, Fernández AF, Fraga MF. A multiomic atlas of the aging hippocampus reveals molecular changes in response to environmental enrichment. Nat Commun 2024; 15:5829. [PMID: 39013876 PMCID: PMC11252340 DOI: 10.1038/s41467-024-49608-z] [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: 09/12/2023] [Accepted: 06/10/2024] [Indexed: 07/18/2024] Open
Abstract
Aging involves the deterioration of organismal function, leading to the emergence of multiple pathologies. Environmental stimuli, including lifestyle, can influence the trajectory of this process and may be used as tools in the pursuit of healthy aging. To evaluate the role of epigenetic mechanisms in this context, we have generated bulk tissue and single cell multi-omic maps of the male mouse dorsal hippocampus in young and old animals exposed to environmental stimulation in the form of enriched environments. We present a molecular atlas of the aging process, highlighting two distinct axes, related to inflammation and to the dysregulation of mRNA metabolism, at the functional RNA and protein level. Additionally, we report the alteration of heterochromatin domains, including the loss of bivalent chromatin and the uncovering of a heterochromatin-switch phenomenon whereby constitutive heterochromatin loss is partially mitigated through gains in facultative heterochromatin. Notably, we observed the multi-omic reversal of a great number of aging-associated alterations in the context of environmental enrichment, which was particularly linked to glial and oligodendrocyte pathways. In conclusion, our work describes the epigenomic landscape of environmental stimulation in the context of aging and reveals how lifestyle intervention can lead to the multi-layered reversal of aging-associated decline.
Collapse
Affiliation(s)
- Raúl F Pérez
- Cancer Epigenetics and Nanomedicine Laboratory, Centro de Investigación en Nanomateriales y Nanotecnología-Consejo Superior de Investigaciones Científicas (CINN-CSIC), Universidad de Oviedo, 33011, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA-FINBA), Universidad de Oviedo, 33011, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, 33003, Oviedo, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain
| | - Patricia Tezanos
- Departamento de Neurociencia Translacional, Instituto Cajal-Consejo Superior de Investigaciones Científicas (IC-CSIC), 28002, Madrid, Spain
- Programa de Doctorado en Neurociencia, Universidad Autónoma de Madrid-Instituto Cajal, 28002, Madrid, Spain
| | - Alfonso Peñarroya
- Cancer Epigenetics and Nanomedicine Laboratory, Centro de Investigación en Nanomateriales y Nanotecnología-Consejo Superior de Investigaciones Científicas (CINN-CSIC), Universidad de Oviedo, 33011, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA-FINBA), Universidad de Oviedo, 33011, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, 33003, Oviedo, Spain
| | - Alejandro González-Ramón
- Laboratory of Functional Epi-Genomics of Aging and Alzheimer's disease, Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), 03550, Alicante, Spain
| | - Rocío G Urdinguio
- Cancer Epigenetics and Nanomedicine Laboratory, Centro de Investigación en Nanomateriales y Nanotecnología-Consejo Superior de Investigaciones Científicas (CINN-CSIC), Universidad de Oviedo, 33011, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA-FINBA), Universidad de Oviedo, 33011, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, 33003, Oviedo, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain
| | - Javier Gancedo-Verdejo
- Cancer Epigenetics and Nanomedicine Laboratory, Centro de Investigación en Nanomateriales y Nanotecnología-Consejo Superior de Investigaciones Científicas (CINN-CSIC), Universidad de Oviedo, 33011, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA-FINBA), Universidad de Oviedo, 33011, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, 33003, Oviedo, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain
| | - Juan Ramón Tejedor
- Cancer Epigenetics and Nanomedicine Laboratory, Centro de Investigación en Nanomateriales y Nanotecnología-Consejo Superior de Investigaciones Científicas (CINN-CSIC), Universidad de Oviedo, 33011, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA-FINBA), Universidad de Oviedo, 33011, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, 33003, Oviedo, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain
| | - Pablo Santamarina-Ojeda
- Cancer Epigenetics and Nanomedicine Laboratory, Centro de Investigación en Nanomateriales y Nanotecnología-Consejo Superior de Investigaciones Científicas (CINN-CSIC), Universidad de Oviedo, 33011, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA-FINBA), Universidad de Oviedo, 33011, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, 33003, Oviedo, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain
| | - Juan José Alba-Linares
- Cancer Epigenetics and Nanomedicine Laboratory, Centro de Investigación en Nanomateriales y Nanotecnología-Consejo Superior de Investigaciones Científicas (CINN-CSIC), Universidad de Oviedo, 33011, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA-FINBA), Universidad de Oviedo, 33011, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, 33003, Oviedo, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain
| | - Lidia Sainz-Ledo
- Cancer Epigenetics and Nanomedicine Laboratory, Centro de Investigación en Nanomateriales y Nanotecnología-Consejo Superior de Investigaciones Científicas (CINN-CSIC), Universidad de Oviedo, 33011, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA-FINBA), Universidad de Oviedo, 33011, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, 33003, Oviedo, Spain
| | - Annalisa Roberti
- Cancer Epigenetics and Nanomedicine Laboratory, Centro de Investigación en Nanomateriales y Nanotecnología-Consejo Superior de Investigaciones Científicas (CINN-CSIC), Universidad de Oviedo, 33011, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA-FINBA), Universidad de Oviedo, 33011, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, 33003, Oviedo, Spain
| | - Virginia López
- Cancer Epigenetics and Nanomedicine Laboratory, Centro de Investigación en Nanomateriales y Nanotecnología-Consejo Superior de Investigaciones Científicas (CINN-CSIC), Universidad de Oviedo, 33011, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA-FINBA), Universidad de Oviedo, 33011, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, 33003, Oviedo, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain
| | - Cristina Mangas
- Cancer Epigenetics and Nanomedicine Laboratory, Centro de Investigación en Nanomateriales y Nanotecnología-Consejo Superior de Investigaciones Científicas (CINN-CSIC), Universidad de Oviedo, 33011, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA-FINBA), Universidad de Oviedo, 33011, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, 33003, Oviedo, Spain
| | - María Moro
- Departamento de Neurociencia Translacional, Instituto Cajal-Consejo Superior de Investigaciones Científicas (IC-CSIC), 28002, Madrid, Spain
| | - Elisa Cintado Reyes
- Departamento de Neurociencia Translacional, Instituto Cajal-Consejo Superior de Investigaciones Científicas (IC-CSIC), 28002, Madrid, Spain
- Programa de Doctorado en Neurociencia, Universidad Autónoma de Madrid-Instituto Cajal, 28002, Madrid, Spain
| | - Pablo Muela Martínez
- Departamento de Neurociencia Translacional, Instituto Cajal-Consejo Superior de Investigaciones Científicas (IC-CSIC), 28002, Madrid, Spain
- Programa de Doctorado en Neurociencia, Universidad Autónoma de Madrid-Instituto Cajal, 28002, Madrid, Spain
| | - Mar Rodríguez-Santamaría
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA-FINBA), Universidad de Oviedo, 33011, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, 33003, Oviedo, Spain
- Bioterio y unidad de imagen preclínica, Universidad de Oviedo, 33006, Oviedo, Spain
| | - Ignacio Ortea
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA-FINBA), Universidad de Oviedo, 33011, Oviedo, Spain
- Proteomics Unit, Centro de Investigación en Nanomateriales y Nanotecnología-Consejo Superior de Investigaciones Científicas (CINN-CSIC), Instituto de Investigación Sanitaria del Principado de Asturias (ISPA-FINBA), 33011, Oviedo, Spain
| | - Ramón Iglesias-Rey
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706, Santiago de Compostela, Spain
| | - Juan Castilla-Silgado
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA-FINBA), Universidad de Oviedo, 33011, Oviedo, Spain
- Departamento de Biología Funcional, Área de Fisiología, Universidad de Oviedo, 33006, Oviedo, Spain
| | - Cristina Tomás-Zapico
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA-FINBA), Universidad de Oviedo, 33011, Oviedo, Spain
- Departamento de Biología Funcional, Área de Fisiología, Universidad de Oviedo, 33006, Oviedo, Spain
| | - Eduardo Iglesias-Gutiérrez
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA-FINBA), Universidad de Oviedo, 33011, Oviedo, Spain
- Departamento de Biología Funcional, Área de Fisiología, Universidad de Oviedo, 33006, Oviedo, Spain
| | - Benjamín Fernández-García
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA-FINBA), Universidad de Oviedo, 33011, Oviedo, Spain
- Departamento de Biología Funcional, Área de Fisiología, Universidad de Oviedo, 33006, Oviedo, Spain
| | - Jose Vicente Sanchez-Mut
- Laboratory of Functional Epi-Genomics of Aging and Alzheimer's disease, Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), 03550, Alicante, Spain
| | - José Luis Trejo
- Departamento de Neurociencia Translacional, Instituto Cajal-Consejo Superior de Investigaciones Científicas (IC-CSIC), 28002, Madrid, Spain
| | - Agustín F Fernández
- Cancer Epigenetics and Nanomedicine Laboratory, Centro de Investigación en Nanomateriales y Nanotecnología-Consejo Superior de Investigaciones Científicas (CINN-CSIC), Universidad de Oviedo, 33011, Oviedo, Spain.
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA-FINBA), Universidad de Oviedo, 33011, Oviedo, Spain.
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, 33003, Oviedo, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain.
| | - Mario F Fraga
- Cancer Epigenetics and Nanomedicine Laboratory, Centro de Investigación en Nanomateriales y Nanotecnología-Consejo Superior de Investigaciones Científicas (CINN-CSIC), Universidad de Oviedo, 33011, Oviedo, Spain.
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA-FINBA), Universidad de Oviedo, 33011, Oviedo, Spain.
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, 33003, Oviedo, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain.
- Departamento de Biología de Organismos y Sistemas, Área de Fisiología Vegetal, Universidad de Oviedo, 33006, Oviedo, Spain.
| |
Collapse
|
8
|
Bénitière F, Duret L, Necsulea A. GTDrift: a resource for exploring the interplay between genetic drift, genomic and transcriptomic characteristics in eukaryotes. NAR Genom Bioinform 2024; 6:lqae064. [PMID: 38867915 PMCID: PMC11167491 DOI: 10.1093/nargab/lqae064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/22/2024] [Accepted: 05/27/2024] [Indexed: 06/14/2024] Open
Abstract
We present GTDrift, a comprehensive data resource that enables explorations of genomic and transcriptomic characteristics alongside proxies of the intensity of genetic drift in individual species. This resource encompasses data for 1506 eukaryotic species, including 1413 animals and 93 green plants, and is organized in three components. The first two components contain approximations of the effective population size, which serve as indicators of the extent of random genetic drift within each species. In the first component, we meticulously investigated public databases to assemble data on life history traits such as longevity, adult body length and body mass for a set of 979 species. The second component includes estimations of the ratio between the rate of non-synonymous substitutions and the rate of synonymous substitutions (dN/dS) in protein-coding sequences for 1324 species. This ratio provides an estimate of the efficiency of natural selection in purging deleterious substitutions. Additionally, we present polymorphism-derived N e estimates for 66 species. The third component encompasses various genomic and transcriptomic characteristics. With this component, we aim to facilitate comparative transcriptomics analyses across species, by providing easy-to-use processed data for more than 16 000 RNA-seq samples across 491 species. These data include intron-centered alternative splicing frequencies, gene expression levels and sequencing depth statistics for each species, obtained with a homogeneous analysis protocol. To enable cross-species comparisons, we provide orthology predictions for conserved single-copy genes based on BUSCO gene sets. To illustrate the possible uses of this database, we identify the most frequently used introns for each gene and we assess how the sequencing depth available for each species affects our power to identify major and minor splice variants.
Collapse
Affiliation(s)
- Florian Bénitière
- Laboratoire de Biométrie et Biologie Évolutive, Université Lyon 1, UMR CNRS 5558, Villeurbanne, France
- Laboratoire d’Écologie des Hydrosystèmes Naturels et Anthropisés, Université Lyon 1, UMR CNRS 5023, Villeurbanne, France
| | - Laurent Duret
- Laboratoire de Biométrie et Biologie Évolutive, Université Lyon 1, UMR CNRS 5558, Villeurbanne, France
| | - Anamaria Necsulea
- Laboratoire de Biométrie et Biologie Évolutive, Université Lyon 1, UMR CNRS 5558, Villeurbanne, France
| |
Collapse
|
9
|
J E T M, P A S, C L, N GK, F GK, R A H. Leopard density and determinants of space use in a farming landscape in South Africa. Sci Rep 2024; 14:10562. [PMID: 38719842 PMCID: PMC11079070 DOI: 10.1038/s41598-024-61013-6] [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: 01/24/2024] [Accepted: 04/30/2024] [Indexed: 05/12/2024] Open
Abstract
Protected areas are traditionally the foundation of conservation strategy, but land not formally protected is of particular importance for the conservation of large carnivores because of their typically wide-ranging nature. In South Africa, leopard (Panthera pardus) population decreases are thought to be occurring in areas of human development and intense negative interactions, but research is biased towards protected areas, with quantitative information on population sizes and trends in non-protected areas severely lacking. Using Spatially Explicit Capture-Recapture and occupancy techniques including 10 environmental and anthropogenic covariates, we analysed camera trap data from commercial farmland in South Africa where negative human-wildlife interactions are reported to be high. Our findings demonstrate that leopards persist at a moderate density (2.21 /100 km2) and exhibit signs of avoidance from areas where lethal control measures are implemented. This suggests leopards have the potential to navigate mixed mosaic landscapes effectively, enhancing their chances of long-term survival and coexistence with humans. Mixed mosaics of agriculture that include crops, game and livestock farming should be encouraged and, providing lethal control is not ubiquitous in the landscape, chains of safer spaces should permit vital landscape connectivity. However, continuing to promote non-lethal mitigation techniques remains vital.
Collapse
Affiliation(s)
- McKaughan J E T
- Department of Anthropology, Durham University, South Road, Durham, DH1 3LE, UK.
- Conservation Ecology Group, Department of Biosciences, Durham University, South Road, Durham, DH1 3LE, UK.
- Primate and Predator Project, Alldays Wildlife and Communities Research Centre, Campfornis Game Farm, Alldays, South Africa.
| | - Stephens P A
- Conservation Ecology Group, Department of Biosciences, Durham University, South Road, Durham, DH1 3LE, UK
| | - Lucas C
- School of Animal, Rural and Environmental Sciences, Nottingham Trent University, Southwell, UK
| | - Guichard-Kruger N
- Primate and Predator Project, Alldays Wildlife and Communities Research Centre, Campfornis Game Farm, Alldays, South Africa
| | - Guichard-Kruger F
- Primate and Predator Project, Alldays Wildlife and Communities Research Centre, Campfornis Game Farm, Alldays, South Africa
| | - Hill R A
- Department of Anthropology, Durham University, South Road, Durham, DH1 3LE, UK
- Primate and Predator Project, Alldays Wildlife and Communities Research Centre, Campfornis Game Farm, Alldays, South Africa
- Department of Biological Sciences, Faculty of Science, Engineering and Agriculture, University of Venda, Private Bag X5050, Thohoyandou, 0950, South Africa
| |
Collapse
|
10
|
Huijsmans TERG, Courtiol A, Van Soom A, Smits K, Rousset F, Wauters J, Hildebrandt TB. Quantifying maternal investment in mammals using allometry. Commun Biol 2024; 7:475. [PMID: 38637653 PMCID: PMC11026411 DOI: 10.1038/s42003-024-06165-x] [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: 10/18/2023] [Accepted: 04/09/2024] [Indexed: 04/20/2024] Open
Abstract
Maternal investment influences the survival and reproduction of both mothers and their progeny and plays a crucial role in understanding individuals' life-history and population ecology. To reveal the complex mechanisms associated with reproduction and investment, it is necessary to examine variations in maternal investment across species. Comparisons across species call for a standardised method to quantify maternal investment, which remained to be developed. This paper addresses this limitation by introducing the maternal investment metric - MI - for mammalian species, established through the allometric scaling of the litter mass at weaning age by the adult mass and investment duration (i.e. gestation + lactation duration) of a species. Using a database encompassing hundreds of mammalian species, we show that the metric is not highly sensitive to the regression method used to fit the allometric relationship or to the proxy used for adult body mass. The comparison of the maternal investment metric between mammalian subclasses and orders reveals strong differences across taxa. For example, our metric confirms that Eutheria have a higher maternal investment than Metatheria. We discuss how further research could use the maternal investment metric as a valuable tool to understand variation in reproductive strategies.
Collapse
Affiliation(s)
- Tim E R G Huijsmans
- Department of Internal Medicine, Reproduction and Population Medicine, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium.
| | - Alexandre Courtiol
- Department of Evolutionary Genetics, Leibniz Institute for Zoo & Wildlife Research, Alfred-Kowalke-Str. 17, 10315, Berlin, Germany
| | - Ann Van Soom
- Department of Internal Medicine, Reproduction and Population Medicine, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium
| | - Katrien Smits
- Department of Internal Medicine, Reproduction and Population Medicine, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium
| | - François Rousset
- Institute of Evolutionary Science of Montpellier, University of Montpellier, CNRS, IRD, campus Triolet, 34095, Montpellier cedex 05, France
| | - Jella Wauters
- Department of Reproduction Biology, Leibniz Institute for Zoo & Wildlife Research, Alfred-Kowalke-Str. 17, 10315, Berlin, Germany
- Laboratory of Integrative Metabolomics, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium
| | - Thomas B Hildebrandt
- Department of Reproduction Management, Leibniz Institute for Zoo & Wildlife Research, Alfred-Kowalke-Str. 17, 10315, Berlin, Germany
- Freie Universität Berlin, Kaiserswerther Str. 16-18, 14195, Berlin, Germany
| |
Collapse
|
11
|
Landsberger T, Amit I, Alon U. Geroprotective interventions converge on gene expression programs of reduced inflammation and restored fatty acid metabolism. GeroScience 2024; 46:1627-1639. [PMID: 37698783 PMCID: PMC10828297 DOI: 10.1007/s11357-023-00915-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 08/15/2023] [Indexed: 09/13/2023] Open
Abstract
Understanding the mechanisms of geroprotective interventions is central to aging research. We compare four prominent interventions: senolysis, caloric restriction, in vivo partial reprogramming, and heterochronic parabiosis. Using published mice transcriptomic data, we juxtapose these interventions against normal aging. We find a gene expression program common to all four interventions, in which inflammation is reduced and several metabolic processes, especially fatty acid metabolism, are increased. Normal aging exhibits the inverse of this signature across multiple organs and tissues. A similar inverse signature arises in three chronic inflammation disease models in a non-aging context, suggesting that the shift in metabolism occurs downstream of inflammation. Chronic inflammation is also shown to accelerate transcriptomic age. We conclude that a core mechanism of geroprotective interventions acts through the reduction of inflammation with downstream effects that restore fatty acid metabolism. This supports the notion of directly targeting genes associated with these pathways to mitigate age-related deterioration.
Collapse
Affiliation(s)
- Tomer Landsberger
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel.
| | - Ido Amit
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel.
| | - Uri Alon
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
12
|
Han JX, Bai Z, Wang RW. Unraveling power-law scaling through exponential cell division dynamics. Biosystems 2024; 238:105190. [PMID: 38492628 DOI: 10.1016/j.biosystems.2024.105190] [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: 12/07/2023] [Revised: 03/11/2024] [Accepted: 03/12/2024] [Indexed: 03/18/2024]
Abstract
A primary objective of biology is the development of universal laws that define how organic form develops and how it evolves as a function of size, both ontogenetically and across evolutionary time. Scaling theory has been essential in reaching this goal by giving a complete perspective point, particularly in illuminating the fundamental biological features produced within scaling exponents defining families of equations. Nonetheless, the theoretical basis of the allometric equation within scaling theory are inadequately explained, particularly when it comes to establishing links between micro-level processes at the cellular level and macro-level phenomena. We proposed an unlimited cell bipartition, resulting in an exponential growth in cell numbers during an individual's lifespan, to bridge this conceptual gap between cellular processes and allometric scaling. The power-law scaling between body mass and organ weight was produced by the synchronous exponential increments and the allometric exponent is rate of logarithmic cell proliferation rate. Substituting organ weight for erythrocyte weight aided in the development of a power-law scaling relationship between body mass and metabolic rate. Furthermore, it is critical to understand how cell size affects the exponent in power-law scaling. We find that a bigger exponent will result from an increase in the average weight of organ cells or a decrease in the average weight of all cells. Furthermore, cell proliferation dynamics showed a complex exponential scaling between body mass and longevity, defying the previously reported power-law scaling. We discovered a quadratic link between longevity and logarithmic body mass. Notably, all of the parameters included in these relationships are explained by indices linked to cell division and embryonic development. This research adds to our understanding of the complex interaction between cellular processes and overarching scaling phenomena in biology.
Collapse
Affiliation(s)
- Jia-Xu Han
- School of Ecology and Environment, Northwestern Polytechnical University, Xi'an 710072, PR China; Zoology Department and Biodiversity Research Centre, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Zhuangdong Bai
- School of Ecology and Environment, Northwestern Polytechnical University, Xi'an 710072, PR China
| | - Rui-Wu Wang
- School of Ecology and Environment, Northwestern Polytechnical University, Xi'an 710072, PR China.
| |
Collapse
|
13
|
Wang X, Jiang Q, Zhang H, He Z, Song Y, Chen Y, Tang N, Zhou Y, Li Y, Antebi A, Wu L, Han JDJ, Shen Y. Tissue-specific profiling of age-dependent miRNAomic changes in Caenorhabditis elegans. Nat Commun 2024; 15:955. [PMID: 38302463 PMCID: PMC10834975 DOI: 10.1038/s41467-024-45249-4] [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: 05/11/2023] [Accepted: 01/18/2024] [Indexed: 02/03/2024] Open
Abstract
Ageing exhibits common and distinct features in various tissues, making it critical to decipher the tissue-specific ageing mechanisms. MiRNAs are essential regulators in ageing and are recently highlighted as a class of intercellular messengers. However, little is known about the tissue-specific transcriptomic changes of miRNAs during ageing. C. elegans is a well-established model organism in ageing research. Here, we profile the age-dependent miRNAomic changes in five isolated worm tissues. Besides the diverse ageing-regulated miRNA expression across tissues, we discover numerous miRNAs in the tissues without their transcription. We further profile miRNAs in the extracellular vesicles and find that worm miRNAs undergo inter-tissue trafficking via these vesicles in an age-dependent manner. Using these datasets, we uncover the interaction between body wall muscle-derived mir-1 and DAF-16/FOXO in the intestine, suggesting mir-1 as a messenger in inter-tissue signalling. Taken together, we systematically investigate worm miRNAs in the somatic tissues and extracellular vesicles during ageing, providing a valuable resource to study tissue-autonomous and nonautonomous functions of miRNAs in ageing.
Collapse
Affiliation(s)
- Xueqing Wang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, 200031, Shanghai, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Quanlong Jiang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology (CQB), Peking University, 102213, Beijing, China
| | - Hongdao Zhang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, 200031, Shanghai, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Zhidong He
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, 200031, Shanghai, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Yuanyuan Song
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, 200031, Shanghai, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Yifan Chen
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, 200031, Shanghai, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Na Tang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, 200031, Shanghai, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Yifei Zhou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, 200031, Shanghai, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Yiping Li
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, 200031, Shanghai, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Adam Antebi
- Max Planck Institute for Biology of Ageing, D-50931, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50674, Cologne, Germany
| | - Ligang Wu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, 200031, Shanghai, China.
- University of Chinese Academy of Sciences, 100049, Beijing, China.
| | - Jing-Dong J Han
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology (CQB), Peking University, 102213, Beijing, China.
| | - Yidong Shen
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, 200031, Shanghai, China.
- University of Chinese Academy of Sciences, 100049, Beijing, China.
| |
Collapse
|
14
|
de Magalhães JP, Abidi Z, dos Santos GA, Avelar RA, Barardo D, Chatsirisupachai K, Clark P, De-Souza EA, Johnson EJ, Lopes I, Novoa G, Senez L, Talay A, Thornton D, To P. Human Ageing Genomic Resources: updates on key databases in ageing research. Nucleic Acids Res 2024; 52:D900-D908. [PMID: 37933854 PMCID: PMC10767973 DOI: 10.1093/nar/gkad927] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/06/2023] [Accepted: 10/09/2023] [Indexed: 11/08/2023] Open
Abstract
Ageing is a complex and multifactorial process. For two decades, the Human Ageing Genomic Resources (HAGR) have aided researchers in the study of various aspects of ageing and its manipulation. Here, we present the key features and recent enhancements of these resources, focusing on its six main databases. One database, GenAge, focuses on genes related to ageing, featuring 307 genes linked to human ageing and 2205 genes associated with longevity and ageing in model organisms. AnAge focuses on ageing, longevity, and life-history across animal species, containing data on 4645 species. DrugAge includes information about 1097 longevity drugs and compounds in model organisms such as mice, rats, flies, worms and yeast. GenDR provides a list of 214 genes associated with the life-extending benefits of dietary restriction in model organisms. CellAge contains a catalogue of 866 genes associated with cellular senescence. The LongevityMap serves as a repository for genetic variants associated with human longevity, encompassing 3144 variants pertaining to 884 genes. Additionally, HAGR provides various tools as well as gene expression signatures of ageing, dietary restriction, and replicative senescence based on meta-analyses. Our databases are integrated, regularly updated, and manually curated by experts. HAGR is freely available online (https://genomics.senescence.info/).
Collapse
Affiliation(s)
- João Pedro de Magalhães
- Genomics of Ageing and Rejuvenation Lab, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2WB, UK
| | - Zoya Abidi
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, UK
| | - Gabriel Arantes dos Santos
- Genomics of Ageing and Rejuvenation Lab, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2WB, UK
| | - Roberto A Avelar
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, UK
| | - Diogo Barardo
- NOVOS Labs, 100 Park Avenue, 16th Fl, New York, NY 10017, USA
| | - Kasit Chatsirisupachai
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, UK
| | - Peter Clark
- Genomics of Ageing and Rejuvenation Lab, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2WB, UK
| | - Evandro A De-Souza
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas 13083-970, SP, Brazil
| | - Emily J Johnson
- Computational Biology Facility, Liverpool Shared Research Facilities, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 7ZB, UK
| | - Inês Lopes
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, UK
| | - Guy Novoa
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, UK
| | - Ludovic Senez
- Genomics of Ageing and Rejuvenation Lab, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2WB, UK
| | - Angelo Talay
- Genomics of Ageing and Rejuvenation Lab, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2WB, UK
| | - Daniel Thornton
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, UK
| | - Paul Ka Po To
- Genomics of Ageing and Rejuvenation Lab, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2WB, UK
| |
Collapse
|
15
|
Lukacsovich D, O’Shea D, Huang H, Zhang W, Young JI, Steven Chen X, Dietrich ST, Kunkle B, Martin ER, Wang L. MIAMI-AD (Methylation in Aging and Methylation in AD): an integrative knowledgebase that facilitates explorations of DNA methylation across sex, aging, and Alzheimer's disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.04.23299412. [PMID: 38105943 PMCID: PMC10723513 DOI: 10.1101/2023.12.04.23299412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disorder with a significant impact on aging populations. DNA methylation (DNAm) alterations have been implicated in both the aging processes and the development of AD. Given that AD affects more women than men, it is also important to explore DNAm changes that occur specifically in each sex. We created MIAMI-AD, a comprehensive knowledge base containing manually curated summary statistics from 97 published tables in 37 studies, all of which included at least 100 participants. MIAMI-AD enables easy browsing, querying, and downloading DNAm associations at multiple levels - at individual CpG, gene, genomic regions, or genome-wide, in one or multiple studies. Moreover, it also offers tools to perform integrative analyses, such as comparing DNAm associations across different phenotypes or tissues, as well as interactive visualizations. Using several use case examples, we demonstrated that MIAMI-AD facilitates our understanding of age-associated CpGs in AD and the sex-specific roles of DNAm in AD. This open-access resource is freely available to the research community, and all the underlying data can be downloaded. MIAMI-AD (https://miami-ad.org/) facilitates integrative explorations to better understand the interplay between DNAm across aging, sex, and AD.
Collapse
Affiliation(s)
- David Lukacsovich
- Division of Biostatistics, Department of Public Health Sciences, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - Deirdre O’Shea
- Department of Neurology, Comprehensive Center for Brain Health, University of Miami Miller School of Medicine, Boca Raton, FL, 33433
| | - Hanchen Huang
- Division of Biostatistics, Department of Public Health Sciences, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - Wei Zhang
- Division of Biostatistics, Department of Public Health Sciences, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - Juan I. Young
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
- John P. Hussman Institute for Human Genomics, the University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - X. Steven Chen
- Division of Biostatistics, Department of Public Health Sciences, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - Sven-Thorsten Dietrich
- John P. Hussman Institute for Human Genomics, the University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Brian Kunkle
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
- John P. Hussman Institute for Human Genomics, the University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Eden R. Martin
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
- John P. Hussman Institute for Human Genomics, the University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Lily Wang
- Division of Biostatistics, Department of Public Health Sciences, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
- John P. Hussman Institute for Human Genomics, the University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
16
|
Wu Z, Feng C, Hu Y, Zhou Y, Li S, Zhang S, Hu Y, Chen Y, Chao H, Ni Q, Chen M. HALD, a human aging and longevity knowledge graph for precision gerontology and geroscience analyses. Sci Data 2023; 10:851. [PMID: 38040715 PMCID: PMC10692171 DOI: 10.1038/s41597-023-02781-0] [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: 05/16/2023] [Accepted: 11/23/2023] [Indexed: 12/03/2023] Open
Abstract
Human aging is a natural and inevitable biological process that leads to an increased risk of aging-related diseases. Developing anti-aging therapies for aging-related diseases requires a comprehensive understanding of the mechanisms and effects of aging and longevity from a multi-modal and multi-faceted perspective. However, most of the relevant knowledge is scattered in the biomedical literature, the volume of which reached 36 million in PubMed. Here, we presented HALD, a text mining-based human aging and longevity dataset of the biomedical knowledge graph from all published literature related to human aging and longevity in PubMed. HALD integrated multiple state-of-the-art natural language processing (NLP) techniques to improve the accuracy and coverage of the knowledge graph for precision gerontology and geroscience analyses. Up to September 2023, HALD had contained 12,227 entities in 10 types (gene, RNA, protein, carbohydrate, lipid, peptide, pharmaceutical preparations, toxin, mutation, and disease), 115,522 relations, 1,855 aging biomarkers, and 525 longevity biomarkers from 339,918 biomedical articles in PubMed. HALD is available at https://bis.zju.edu.cn/hald .
Collapse
Affiliation(s)
- Zexu Wu
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Cong Feng
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
- The First Affiliated Hospital, Zhejiang University School of Medicine; Institute of Hematology, Zhejiang University, Hangzhou, 310058, China
| | - Yanshi Hu
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yincong Zhou
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
- Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, 314400, China
| | - Sida Li
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Shilong Zhang
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yueming Hu
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yuhao Chen
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Haoyu Chao
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Qingyang Ni
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Ming Chen
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China.
- The First Affiliated Hospital, Zhejiang University School of Medicine; Institute of Hematology, Zhejiang University, Hangzhou, 310058, China.
- Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, 314400, China.
| |
Collapse
|
17
|
He Z, He W, Hu C, Liao J, Deng W, Sun H, Huang Q, Chen W, Zhang L, Liu M, Dong J. Cross-species comparison illuminates the importance of iron homeostasis for splenic anti-immunosenescence. Aging Cell 2023; 22:e13982. [PMID: 37681451 PMCID: PMC10652311 DOI: 10.1111/acel.13982] [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: 02/20/2023] [Revised: 08/21/2023] [Accepted: 08/23/2023] [Indexed: 09/09/2023] Open
Abstract
Although immunosenescence may result in increased morbidity and mortality, many mammals have evolved effective immune coping strategies to extend their lifespans. Thus, the immune systems of long-lived mammals present unique models to study healthy longevity. To identify the molecular clues of anti-immunosenescence, we first built high-quality reference genome for a long-lived myotis bat, and then compared three long-lived mammals (i.e., bat, naked mole rat, and human) versus the short-lived mammal, mouse, in splenic immune cells at single-cell resolution. A close relationship between B:T cell ratio and immunosenescence was detected, as B:T cell ratio was much higher in mouse than long-lived mammals and significantly increased during aging. Importantly, we identified several iron-related genes that could resist immunosenescence changes, especially the iron chaperon, PCBP1, which was upregulated in long-lived mammals but dramatically downregulated during aging in all splenic immune cell types. Supportively, immune cells of mouse spleens contained more free iron than those of bat spleens, suggesting higher level of ROS-induced damage in mouse. PCBP1 downregulation during aging was also detected in hepatic but not pulmonary immune cells, which is consistent with the crucial roles of spleen and liver in organismal iron recycling. Furthermore, PCBP1 perturbation in immune cell lines would result in cellular iron dyshomeostasis and senescence. Finally, we identified two transcription factors that could regulate PCBP1 during aging. Together, our findings highlight the importance of iron homeostasis in splenic anti-immunosenescence, and provide unique insight for improving human healthspan.
Collapse
Affiliation(s)
- Ziqing He
- GMU‐GIBH Joint School of Life Sciences, The Guangdong‐Hong Kong‐Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou National LaboratoryGuangzhou Medical UniversityGuangzhouChina
- Faculty of Health SciencesUniversity of MacauMacauChina
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory)GuangzhouChina
| | - Weiya He
- GMU‐GIBH Joint School of Life Sciences, The Guangdong‐Hong Kong‐Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou National LaboratoryGuangzhou Medical UniversityGuangzhouChina
- Faculty of Health SciencesUniversity of MacauMacauChina
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory)GuangzhouChina
| | - Chuanxia Hu
- GMU‐GIBH Joint School of Life Sciences, The Guangdong‐Hong Kong‐Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou National LaboratoryGuangzhou Medical UniversityGuangzhouChina
| | - Jiayu Liao
- GMU‐GIBH Joint School of Life Sciences, The Guangdong‐Hong Kong‐Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou National LaboratoryGuangzhou Medical UniversityGuangzhouChina
| | - Wenjun Deng
- GMU‐GIBH Joint School of Life Sciences, The Guangdong‐Hong Kong‐Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou National LaboratoryGuangzhou Medical UniversityGuangzhouChina
| | - Haijian Sun
- GMU‐GIBH Joint School of Life Sciences, The Guangdong‐Hong Kong‐Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou National LaboratoryGuangzhou Medical UniversityGuangzhouChina
- Faculty of Health SciencesUniversity of MacauMacauChina
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory)GuangzhouChina
| | - Qingpei Huang
- GMU‐GIBH Joint School of Life Sciences, The Guangdong‐Hong Kong‐Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou National LaboratoryGuangzhou Medical UniversityGuangzhouChina
| | - Weilue Chen
- GMU‐GIBH Joint School of Life Sciences, The Guangdong‐Hong Kong‐Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou National LaboratoryGuangzhou Medical UniversityGuangzhouChina
| | - Libiao Zhang
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and UtilizationInstitute of Zoology, Guangdong Academy of SciencesGuangzhouChina
| | - Meiling Liu
- GMU‐GIBH Joint School of Life Sciences, The Guangdong‐Hong Kong‐Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou National LaboratoryGuangzhou Medical UniversityGuangzhouChina
| | - Ji Dong
- GMU‐GIBH Joint School of Life Sciences, The Guangdong‐Hong Kong‐Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou National LaboratoryGuangzhou Medical UniversityGuangzhouChina
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory)GuangzhouChina
| |
Collapse
|
18
|
Tidière M, Colchero F, Staerk J, Adkesson MJ, Andersen DH, Bland L, Böye M, Brando S, Clegg I, Cubaynes S, Cutting A, De Man D, Derocher AE, Dorsey C, Elgar W, Gaglione E, Anderson Hansen K, Jungheim A, Kok J, Laule G, Goya AL, Miller L, Monreal-Pawlowsky T, Mucha K, Owen MA, Petersen SD, Pilfold N, Richardson D, Richardson ES, Sabo D, Sato N, Shellabarger W, Skovlund CR, Tomisawa K, Trautwein SE, Van Bonn W, Van Elk C, Von Fersen L, Wahlberg M, Zhang P, Zhang X, Conde DA. Survival improvements of marine mammals in zoological institutions mirror historical advances in human longevity. Proc Biol Sci 2023; 290:20231895. [PMID: 37848064 PMCID: PMC10581765 DOI: 10.1098/rspb.2023.1895] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 09/21/2023] [Indexed: 10/19/2023] Open
Abstract
An intense public debate has fuelled governmental bans on marine mammals held in zoological institutions. The debate rests on the assumption that survival in zoological institutions has been and remains lower than in the wild, albeit the scientific evidence in support of this notion is equivocal. Here, we used statistical methods previously applied to assess historical improvements in human lifespan and data on 8864 individuals of four marine mammal species (harbour seal, Phoca vitulina; California sea lion, Zalophus californianus; polar bear, Ursus maritimus; common bottlenose dolphin, Tursiops truncatus) held in zoos from 1829 to 2020. We found that life expectancy increased up to 3.40 times, and first-year mortality declined up to 31%, during the last century in zoos. Moreover, the life expectancy of animals in zoos is currently 1.65-3.55 times longer than their wild counterparts. Like humans, these improvements have occurred concurrently with advances in management practices, crucial for population welfare. Science-based decisions will help effective legislative changes and ensure better implementation of animal care.
Collapse
Affiliation(s)
- Morgane Tidière
- Interdisciplinary Centre on Population Dynamics (CPop), University of Southern Denmark, Odense, Denmark
- Department of Biology, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark
- Conservation and Science Department, Species360, 7900 International Drive, Suite 300, Minneapolis, MN 55425, USA
| | - Fernando Colchero
- Interdisciplinary Centre on Population Dynamics (CPop), University of Southern Denmark, Odense, Denmark
- Department of Mathematics and Computer Science, University of Southern Denmark, Odense, Denmark
- Department of Primate Behavior and Evolution, Max Planck Institute for Evolutionary Anthropology, Deutscher Pl. 6, 04103 Leipzig, Germany
| | - Johanna Staerk
- Interdisciplinary Centre on Population Dynamics (CPop), University of Southern Denmark, Odense, Denmark
- Department of Biology, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark
- Conservation and Science Department, Species360, 7900 International Drive, Suite 300, Minneapolis, MN 55425, USA
| | | | - Ditte H. Andersen
- Department of Biology, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark
| | - Lucie Bland
- Conservation and Science Department, Species360, 7900 International Drive, Suite 300, Minneapolis, MN 55425, USA
- Eureka Publishing, Thornbury, Australia
| | - Martin Böye
- Centre de Recherche et d'Etude pour l'Animal Sauvage, Planète Sauvage, 44710 Port Saint Pere, France
| | - Sabrina Brando
- AnimalConcepts, PO Box 378, 03725 Teulada, Alicante, Spain
| | - Isabella Clegg
- Animal Welfare Expertise, The Knoll, Woodlands, Combe Martin, EX34 0ATLittleton Manor, Winchester SO22 6QU, UK
| | - Sarah Cubaynes
- CEFE, Univ Montpellier, CNRS, EPHE-PSL University, IRD, Montpellier, France
| | - Amy Cutting
- Polar Bear International, PO Box 3008, Bozeman, MT, USA
| | - Danny De Man
- European Association of Zoos and Aquaria (EAZA), Plantage Middelaan 45, 1018-DC Amsterdam, The Netherlands
| | - Andrew E. Derocher
- Department of Biological Sciences, University of Alberta; Edmonton, Alberta, Canada T6G 2E9
| | - Candice Dorsey
- Association of Zoos and Aquariums, 8403 Colesville Road Ste 710, Silver Spring, MD 20910, USA
| | - William Elgar
- Zoo Miami, 12400 SW 152 Street, Miami, FL 33177, USA
| | - Eric Gaglione
- Georgia Aquarium, 225 Baker Street, Atlanta, GA 30313, USA
| | - Kirstin Anderson Hansen
- Department of Biology, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark
- Marine Biological Research Center, University of Southern Denmark, Hindsholmvej 11, 5300 Kerteminde, Denmark
| | - Allison Jungheim
- Como Park Zoo and Conservatory, 1225 Estabrook Dr., Saint Paul, MN 55103, USA
| | - José Kok
- Ouwehands Zoo, Grebbeweg 111, 3911 AV Rhenen, The Netherlands
| | - Gail Laule
- Mandai Wildlife Group, 80 Mandai Lake Road, Singapore 729826
| | | | - Lance Miller
- Chicago Zoological Society, Brookfield Zoo, Brookfield, IL, USA
| | | | - Katelyn Mucha
- Conservation and Science Department, Species360, 7900 International Drive, Suite 300, Minneapolis, MN 55425, USA
| | - Megan A. Owen
- San Diego Zoo Wildlife Alliance, 15600 San Pasqual Valley Rd., Escondido, CA, USA
| | | | - Nicholas Pilfold
- San Diego Zoo Wildlife Alliance, 15600 San Pasqual Valley Rd., Escondido, CA, USA
| | - Douglas Richardson
- Zoological Consultancy Ltd, Columba Cottage, Mill Rd, Kingussie PH21 1LF, UK
- EAZA Polar Bear EEP, Amsterdam, Netherlands
| | - Evan S. Richardson
- Environment and Climate Change Canada, Unit 150–234 Donald Street, Winnipeg, Manitoba R3C 1M8, Canada
| | - Devon Sabo
- Columbus Zoo and Aquarium, 4850 W. Powell Road, PO Box 400, Powell, OH 43065-0400, USA
| | - Nobutaka Sato
- Asahiyama Zoological Park, Kuranuma, Higasiasahikawacho, Asahikawa city, Japan
| | | | - Cecilie R. Skovlund
- Conservation, Copenhagen Zoo, Roskildevej 38, 2000 Frederiksberg, Denmark
- Section of Animal Welfare and Disease Control, Department of Veterinary and Animal Sciences, University of Copenhagen, Grønnegårdsvej 8, 1870 Frederiksberg, Denmark
| | - Kanako Tomisawa
- Omuta City Zoo, 163 Showa-machi, Omuta, Fukuoka 836-0871, Japan
| | - Sandra E. Trautwein
- Conservation and Science Department, Species360, 7900 International Drive, Suite 300, Minneapolis, MN 55425, USA
| | - William Van Bonn
- A. Watson Armour III, Center for Animal Health and Welfare, Animal Care and Science Division, John G. Shedd Aquarium, Chicago, IL 60605, USA
| | - Cornelis Van Elk
- Independent practitioner, Arendsweg 98, Enschede 7544RM, The Netherlands
| | | | - Magnus Wahlberg
- Department of Biology, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark
- Marine Biological Research Center, University of Southern Denmark, Hindsholmvej 11, 5300 Kerteminde, Denmark
| | - Peijun Zhang
- Mammal and Marine Bioacoustics Laboratory Institute of Deep-sea Science and Engineering, Chinese Academy of Sciences, Sanya 572000, People's Republic of China
| | - Xianfeng Zhang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China
| | - Dalia A. Conde
- Interdisciplinary Centre on Population Dynamics (CPop), University of Southern Denmark, Odense, Denmark
- Department of Biology, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark
| |
Collapse
|
19
|
Danis T, Rokas A. The evolution of gestation length in eutherian mammals. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.22.563491. [PMID: 37961105 PMCID: PMC10634735 DOI: 10.1101/2023.10.22.563491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Gestation length, or the duration of pregnancy, is a critical component of mammalian reproductive biology1. Eutherian mammals exhibit striking variation in their gestation lengths2-5, which has traditionally been linked to and allometrically scales with variation in other life history traits, including body mass and lifespan5-8. How the phenotypic landscape of gestation length variation, including its associations with body mass and lifespan variation, changed over mammalian evolution remains unknown. Phylogeny-informed analyses of 845 representative extant eutherian mammals showed that gestation length variation substantially differed in both whether and how strongly it was associated with body mass and lifespan across mammalian clades. For example, gestation length variation in Chiroptera and Cetacea was not associated with lifespan or body mass but was strongly associated only with body mass in Carnivora. We also identified 52 adaptive shifts in gestation length variation across the mammal phylogeny and 14 adaptive shifts when considering all three life history traits; the placements of six adaptive shifts are common in the two analyses. Notably, two of these shifts occurred at the roots of Cetacea and Pinnipedia, respectively, coinciding with the transition of these clades to the marine environment. The varying dynamics of the phenotypic landscape of gestation length, coupled with the varying patterns of associations between gestation length and two other major life history traits, raise the hypothesis that evolutionary constraints on gestation length have varied substantially across mammalian phylogeny. This variation in constraints implies that the genetic architecture of gestation length differs between mammal clades.
Collapse
Affiliation(s)
- Thodoris Danis
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37235, USA
- Evolutionary Studies Initiative, Vanderbilt University, Nashville, TN 37235, USA
| | - Antonis Rokas
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37235, USA
- Evolutionary Studies Initiative, Vanderbilt University, Nashville, TN 37235, USA
| |
Collapse
|
20
|
Huang Z, Jiang C, Gu J, Uvizl M, Power S, Douglas D, Kacprzyk J. Duplications of Human Longevity-Associated Genes Across Placental Mammals. Genome Biol Evol 2023; 15:evad186. [PMID: 37831410 PMCID: PMC10588791 DOI: 10.1093/gbe/evad186] [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: 05/02/2023] [Revised: 07/31/2023] [Accepted: 10/09/2023] [Indexed: 10/14/2023] Open
Abstract
Natural selection has shaped a wide range of lifespans across mammals, with a few long-lived species showing negligible signs of ageing. Approaches used to elucidate the genetic mechanisms underlying mammalian longevity usually involve phylogenetic selection tests on candidate genes, detections of convergent amino acid changes in long-lived lineages, analyses of differential gene expression between age cohorts or species, and measurements of age-related epigenetic changes. However, the link between gene duplication and evolution of mammalian longevity has not been widely investigated. Here, we explored the association between gene duplication and mammalian lifespan by analyzing 287 human longevity-associated genes across 37 placental mammals. We estimated that the expansion rate of these genes is eight times higher than their contraction rate across these 37 species. Using phylogenetic approaches, we identified 43 genes whose duplication levels are significantly correlated with longevity quotients (False Discovery Rate (FDR) < 0.05). In particular, the strong correlation observed for four genes (CREBBP, PIK3R1, HELLS, FOXM1) appears to be driven mainly by their high duplication levels in two ageing extremists, the naked mole rat (Heterocephalus glaber) and the greater mouse-eared bat (Myotis myotis). Further sequence and expression analyses suggest that the gene PIK3R1 may have undergone a convergent duplication event, whereby the similar region of its coding sequence was independently duplicated multiple times in both of these long-lived species. Collectively, this study identified several candidate genes whose duplications may underlie the extreme longevity in mammals, and highlighted the potential role of gene duplication in the evolution of mammalian long lifespans.
Collapse
Affiliation(s)
- Zixia Huang
- School of Biology and Environmental Science, University College Dublin, Dublin, Ireland
| | - Chongyi Jiang
- Institute of Ecology and Evolution, Friedrich Schiller University, Jena, Germany
| | - Jiayun Gu
- School of Biology and Environmental Science, University College Dublin, Dublin, Ireland
| | - Marek Uvizl
- Department of Zoology, National Museum, Prague, Czech Republic
- Department of Zoology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Sarahjane Power
- School of Biology and Environmental Science, University College Dublin, Dublin, Ireland
| | - Declan Douglas
- School of Biology and Environmental Science, University College Dublin, Dublin, Ireland
| | - Joanna Kacprzyk
- School of Biology and Environmental Science, University College Dublin, Dublin, Ireland
| |
Collapse
|
21
|
Liu Y, Du X, Li G, Liu Y, Li S. Life-History and Ecological Correlates of Egg and Clutch Mass Variation in Sympatric Bird Species at High Altitude. BIOLOGY 2023; 12:1303. [PMID: 37887013 PMCID: PMC10604263 DOI: 10.3390/biology12101303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/19/2023] [Accepted: 09/25/2023] [Indexed: 10/28/2023]
Abstract
The variation in egg and clutch mass in sympatric species at high altitudes is poorly understood, and the potential causes of variation are rarely investigated. This study aimed to describe the interspecific variation in avian egg and clutch mass among 22 sympatric bird species at an altitude of 3430 m. Our objective was to reduce potential confounding effects of biotic/abiotic factors and investigated hypotheses concerning allometry, clutch size, parental care, nest predation, and lifespan as possible correlates and explanations for the observed variation. Our findings indicated that both egg and clutch mass evolve with body mass across species. We found that egg mass variation was not explained by clutch size when controlling for allometric effects, which contrasts the "egg mass vs. clutch size trade-off" hypothesis. Additionally, we found that clutch mass was positively associated with parental care (reflected by development period) but negatively associated with predation rate. By substituting egg mass and clutch size into the models, we found that clutch size was significantly correlated with parental care, predation rate, and lifespan, while egg mass was only significantly associated with development period. Overall, these findings support life-history theories suggesting that reduced clutch size or mass is associated with a higher risk of predation, reduced parental care, but longer adult lifespan. Interestingly, our results indicate that clutch size has a greater influence on these factors compared to egg mass. This could be attributed to the fact that smaller clutch sizes result in a more notable decrease in energetic allocation, as they require a reduced effort in terms of offspring production, incubation, and feeding, as opposed to solely reducing egg size. These findings contribute to the growing evidence that life-history and ecological traits correlate with egg and clutch mass variation in sympatric species. However, further research is needed to explore the potential evolutionary causes underlying these patterns.
Collapse
Affiliation(s)
| | | | | | - Yingbao Liu
- College of Life Sciences, Yangtze University, Jingzhou 434025, China
| | - Shaobin Li
- College of Life Sciences, Yangtze University, Jingzhou 434025, China
| |
Collapse
|
22
|
Liu W, Zhu P, Li M, Li Z, Yu Y, Liu G, Du J, Wang X, Yang J, Tian R, Seim I, Kaya A, Li M, Li M, Gladyshev VN, Zhou X. Large-scale across species transcriptomic analysis identifies genetic selection signatures associated with longevity in mammals. EMBO J 2023; 42:e112740. [PMID: 37427458 PMCID: PMC10476176 DOI: 10.15252/embj.2022112740] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 05/12/2023] [Accepted: 05/22/2023] [Indexed: 07/11/2023] Open
Abstract
Lifespan varies significantly among mammals, with more than 100-fold difference between the shortest and longest living species. This natural difference may uncover the evolutionary forces and molecular features that define longevity. To understand the relationship between gene expression variation and longevity, we conducted a comparative transcriptomics analysis of liver, kidney, and brain tissues of 103 mammalian species. We found that few genes exhibit common expression patterns with longevity in the three organs analyzed. However, pathways related to translation fidelity, such as nonsense-mediated decay and eukaryotic translation elongation, correlated with longevity across mammals. Analyses of selection pressure found that selection intensity related to the direction of longevity-correlated genes is inconsistent across organs. Furthermore, expression of methionine restriction-related genes correlated with longevity and was under strong selection in long-lived mammals, suggesting that a common strategy is utilized by natural selection and artificial intervention to control lifespan. Our results indicate that lifespan regulation via gene expression is driven through polygenic and indirect natural selection.
Collapse
Affiliation(s)
- Weiqiang Liu
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of ZoologyChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Pingfen Zhu
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of ZoologyChinese Academy of SciencesBeijingChina
| | - Meng Li
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of ZoologyChinese Academy of SciencesBeijingChina
| | - Zihao Li
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of ZoologyChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Yang Yu
- School of Life SciencesUniversity of Science and Technology of ChinaAnhuiChina
| | - Gaoming Liu
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of ZoologyChinese Academy of SciencesBeijingChina
| | - Juan Du
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of ZoologyChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Xiao Wang
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of ZoologyChinese Academy of SciencesBeijingChina
| | - Jing Yang
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of ZoologyChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Ran Tian
- Integrative Biology Laboratory, College of Life SciencesNanjing Normal UniversityNanjingChina
| | - Inge Seim
- Integrative Biology Laboratory, College of Life SciencesNanjing Normal UniversityNanjingChina
- School of Biology and Environmental ScienceQueensland University of TechnologyBrisbaneQLDAustralia
| | - Alaattin Kaya
- Department of BiologyVirginia Commonwealth UniversityRichmondVAUSA
| | - Mingzhou Li
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural UniversityChengduChina
| | - Ming Li
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of ZoologyChinese Academy of SciencesBeijingChina
| | - Vadim N Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
| | - Xuming Zhou
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of ZoologyChinese Academy of SciencesBeijingChina
| |
Collapse
|
23
|
Bianucci G, Lambert O, Urbina M, Merella M, Collareta A, Bennion R, Salas-Gismondi R, Benites-Palomino A, Post K, de Muizon C, Bosio G, Di Celma C, Malinverno E, Pierantoni PP, Villa IM, Amson E. A heavyweight early whale pushes the boundaries of vertebrate morphology. Nature 2023; 620:824-829. [PMID: 37532931 DOI: 10.1038/s41586-023-06381-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 06/28/2023] [Indexed: 08/04/2023]
Abstract
The fossil record of cetaceans documents how terrestrial animals acquired extreme adaptations and transitioned to a fully aquatic lifestyle1,2. In whales, this is associated with a substantial increase in maximum body size. Although an elongate body was acquired early in cetacean evolution3, the maximum body mass of baleen whales reflects a recent diversification that culminated in the blue whale4. More generally, hitherto known gigantism among aquatic tetrapods evolved within pelagic, active swimmers. Here we describe Perucetus colossus-a basilosaurid whale from the middle Eocene epoch of Peru. It displays, to our knowledge, the highest degree of bone mass increase known to date, an adaptation associated with shallow diving5. The estimated skeletal mass of P. colossus exceeds that of any known mammal or aquatic vertebrate. We show that the bone structure specializations of aquatic mammals are reflected in the scaling of skeletal fraction (skeletal mass versus whole-body mass) across the entire disparity of amniotes. We use the skeletal fraction to estimate the body mass of P. colossus, which proves to be a contender for the title of heaviest animal on record. Cetacean peak body mass had already been reached around 30 million years before previously assumed, in a coastal context in which primary productivity was particularly high.
Collapse
Affiliation(s)
- Giovanni Bianucci
- Dipartimento di Scienze della Terra, Università di Pisa, Pisa, Italy
| | - Olivier Lambert
- D.O. Terre et Histoire de la Vie, Institut Royal des Sciences Naturelles de Belgique, Brussels, Belgium
| | - Mario Urbina
- Departamento de Paleontología de Vertebrados, Museo de Historia Natural-Universidad Nacional Mayor de San Marcos, Lima, Perú
| | - Marco Merella
- Dipartimento di Scienze della Terra, Università di Pisa, Pisa, Italy
| | - Alberto Collareta
- Dipartimento di Scienze della Terra, Università di Pisa, Pisa, Italy
| | - Rebecca Bennion
- D.O. Terre et Histoire de la Vie, Institut Royal des Sciences Naturelles de Belgique, Brussels, Belgium
- Evolution & Diversity Dynamics Lab, UR Geology, Université de Liège, Liège, Belgium
| | - Rodolfo Salas-Gismondi
- Departamento de Paleontología de Vertebrados, Museo de Historia Natural-Universidad Nacional Mayor de San Marcos, Lima, Perú
- Facultad de Ciencias y Filosofía/Centro de Investigación para el Desarrollo Integral y Sostenible, Laboratorios de Investigación y Desarrollo, Universitad Peruana Cayetano Heredia Lima, Lima, Perú
| | - Aldo Benites-Palomino
- Departamento de Paleontología de Vertebrados, Museo de Historia Natural-Universidad Nacional Mayor de San Marcos, Lima, Perú
- Department of Paleontology, University of Zurich, Zurich, Switzerland
| | - Klaas Post
- Natuurhistorisch Museum Rotterdam, Rotterdam, The Netherlands
| | - Christian de Muizon
- Département Origines et Évolution, CR2P (CNRS, MNHN, Sorbonne Université), Muséum National d'Histoire Naturelle, Paris, France
| | - Giulia Bosio
- Dipartimento di Scienze dell'Ambiente e della Terra, Università degli Studi di Milano-Bicocca, Milano, Italy
| | - Claudio Di Celma
- School of Science and Technology, University of Camerino, Camerino, Italy
| | - Elisa Malinverno
- Dipartimento di Scienze dell'Ambiente e della Terra, Università degli Studi di Milano-Bicocca, Milano, Italy
| | | | | | - Eli Amson
- Staatliches Museum für Naturkunde Stuttgart, Stuttgart, Germany.
| |
Collapse
|
24
|
Jacobs PJ, Hart DW, Merchant HN, Voigt C, Bennett NC. The Evolution and Ecology of Oxidative and Antioxidant Status: A Comparative Approach in African Mole-Rats. Antioxidants (Basel) 2023; 12:1486. [PMID: 37627481 PMCID: PMC10451868 DOI: 10.3390/antiox12081486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/18/2023] [Accepted: 07/20/2023] [Indexed: 08/27/2023] Open
Abstract
The naked mole-rat of the family Bathyergidae has been the showpiece for ageing research as they contradict the traditional understanding of the oxidative stress theory of ageing. Some other bathyergids also possess increased lifespans, but there has been a remarkable lack of comparison between species within the family Bathyergidae. This study set out to investigate how plasma oxidative markers (total oxidant status (TOS), total antioxidant capacity (TAC), and the oxidative stress index (OSI)) differ between five species and three subspecies of bathyergids, differing in their maximum lifespan potential (MLSP), resting metabolic rate, aridity index (AI), and sociality. We also investigated how oxidative markers may differ between captive and wild-caught mole-rats. Our results reveal that increased TOS, TAC, and OSI are associated with increased MLSP. This pattern is more prevalent in the social-living species than the solitary-living species. We also found that oxidative variables decreased with an increasing AI and that wild-caught individuals typically have higher antioxidants. We speculate that the correlation between higher oxidative markers and MLSP is due to the hypoxia-tolerance of the mole-rats investigated. Hormesis (the biphasic response to oxidative stress promoting protection) is a likely mechanism behind the increased oxidative markers observed and promotes longevity in some members of the Bathyergidae family.
Collapse
Affiliation(s)
- Paul. J. Jacobs
- Department of Zoology and Entomology, University of Pretoria, Pretoria 0002, South Africa;
| | - Daniel W. Hart
- Department of Zoology and Entomology, University of Pretoria, Pretoria 0002, South Africa;
| | - Hana N. Merchant
- Department of Biological Sciences, School of Life and Environmental Sciences, Royal Holloway University of London, Egham Hill, Egham, Surrey TW20 0EX, UK;
| | - Cornelia Voigt
- Mammal Research Institute, Department of Zoology and Entomology, University of Pretoria, Pretoria 0002, South Africa; (C.V.); (N.C.B.)
| | - Nigel C. Bennett
- Mammal Research Institute, Department of Zoology and Entomology, University of Pretoria, Pretoria 0002, South Africa; (C.V.); (N.C.B.)
| |
Collapse
|
25
|
Dujon AM, Vincze O, Lemaitre JF, Alix-Panabières C, Pujol P, Giraudeau M, Ujvari B, Thomas F. The effect of placentation type, litter size, lactation and gestation length on cancer risk in mammals. Proc Biol Sci 2023; 290:20230940. [PMID: 37357861 PMCID: PMC10291710 DOI: 10.1098/rspb.2023.0940] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 06/01/2023] [Indexed: 06/27/2023] Open
Abstract
Reproduction is a central activity for all living organisms but is also associated with a diversity of costs that are detrimental for survival. Until recently, the cost of cancer as a selective force has been poorly considered. Considering 191 mammal species, we found cancer mortality was more likely to be detected in species having large, rather than low, litter sizes and long lactation lengths regardless of the placentation types. However, increasing litter size and gestation length are not per se associated with an enhanced cancer mortality risk. Contrary to basic theoretical expectations, the species with the highest cancer mortality were not those with the most invasive (i.e. haemochorial) placentation, but those with a moderately invasive (i.e. endotheliochorial) one. Overall, these results suggest that (i) high reproductive efforts favour oncogenic processes' dynamics, presumably because of trade-offs between allocation in reproduction effort and anti-cancer defences, (ii) cancer defence mechanisms in animals are most often adjusted to align reproductive lifespan, and (iii) malignant cells co-opt existing molecular and physiological pathways for placentation, but species with the most invasive placentation have also selected for potent barriers against lethal cancers. This work suggests that the logic of Peto's paradox seems to be applicable to other traits that promote tumorigenesis.
Collapse
Affiliation(s)
- Antoine M. Dujon
- Geelong, School of Life and Environmental Sciences, Centre for Integrative Ecology, Deakin University, Waurn Ponds, Victoria 3216, Australia
- CREEC/CANECEV (CREES), MIVEGEC, IRD 224–CNRS 5290–Université de Montpellier, Montpellier, France
| | - Orsolya Vincze
- Institute of Aquatic Ecology, Centre for Ecological Research, Debrecen, Hungary
- Evolutionary Ecology Group, Hungarian Department of Biology and Ecology, Babes-Bolyai University, Cluj-Napoca, Romania
| | - Jean-François Lemaitre
- CNRS, UMR 5558, Laboratoire de Biométrie et Biologie Evolutive, Université de Lyon, Université Lyon 1, Villeurbanne, France
| | - Catherine Alix-Panabières
- CREEC/CANECEV (CREES), MIVEGEC, IRD 224–CNRS 5290–Université de Montpellier, Montpellier, France
- Laboratory of Rare Human Circulating Cells (LCCRH), University Hospital of Montpellier, Montpellier, France
| | - Pascal Pujol
- CREEC/CANECEV (CREES), MIVEGEC, IRD 224–CNRS 5290–Université de Montpellier, Montpellier, France
- Centre Hospitalier Universitaire Arnaud de Villeneuve, Montpellier, France
| | - Mathieu Giraudeau
- Littoral Environnement Et Sociétés (LIENSs), UMR 7266,CNRS- La Rochelle Université, La Rochelle, France
| | - Beata Ujvari
- Geelong, School of Life and Environmental Sciences, Centre for Integrative Ecology, Deakin University, Waurn Ponds, Victoria 3216, Australia
- CREEC/CANECEV (CREES), MIVEGEC, IRD 224–CNRS 5290–Université de Montpellier, Montpellier, France
| | - Frédéric Thomas
- CREEC/CANECEV (CREES), MIVEGEC, IRD 224–CNRS 5290–Université de Montpellier, Montpellier, France
| |
Collapse
|
26
|
Tyshkovskiy A, Ma S, Shindyapina AV, Tikhonov S, Lee SG, Bozaykut P, Castro JP, Seluanov A, Schork NJ, Gorbunova V, Dmitriev SE, Miller RA, Gladyshev VN. Distinct longevity mechanisms across and within species and their association with aging. Cell 2023; 186:2929-2949.e20. [PMID: 37269831 PMCID: PMC11192172 DOI: 10.1016/j.cell.2023.05.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/29/2022] [Accepted: 05/02/2023] [Indexed: 06/05/2023]
Abstract
Lifespan varies within and across species, but the general principles of its control remain unclear. Here, we conducted multi-tissue RNA-seq analyses across 41 mammalian species, identifying longevity signatures and examining their relationship with transcriptomic biomarkers of aging and established lifespan-extending interventions. An integrative analysis uncovered shared longevity mechanisms within and across species, including downregulated Igf1 and upregulated mitochondrial translation genes, and unique features, such as distinct regulation of the innate immune response and cellular respiration. Signatures of long-lived species were positively correlated with age-related changes and enriched for evolutionarily ancient essential genes, involved in proteolysis and PI3K-Akt signaling. Conversely, lifespan-extending interventions counteracted aging patterns and affected younger, mutable genes enriched for energy metabolism. The identified biomarkers revealed longevity interventions, including KU0063794, which extended mouse lifespan and healthspan. Overall, this study uncovers universal and distinct strategies of lifespan regulation within and across species and provides tools for discovering longevity interventions.
Collapse
Affiliation(s)
- Alexander Tyshkovskiy
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow 119234, Russia
| | - Siming Ma
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Anastasia V Shindyapina
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Stanislav Tikhonov
- Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow 119234, Russia
| | - Sang-Goo Lee
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Perinur Bozaykut
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey
| | - José P Castro
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; Aging and Aneuploidy Laboratory, IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Andrei Seluanov
- Departments of Biology and Medicine, University of Rochester, Rochester, NY, USA
| | - Nicholas J Schork
- Quantitative Medicine and Systems Biology Division, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Vera Gorbunova
- Departments of Biology and Medicine, University of Rochester, Rochester, NY, USA
| | - Sergey E Dmitriev
- Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow 119234, Russia
| | - Richard A Miller
- Department of Pathology and Geriatrics Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Vadim N Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Broad Institute, Cambridge, MA, USA.
| |
Collapse
|
27
|
de Sousa AA, Rigby Dames BA, Graff EC, Mohamedelhassan R, Vassilopoulos T, Charvet CJ. Going beyond established model systems of Alzheimer's disease: companion animals provide novel insights into the neurobiology of aging. Commun Biol 2023; 6:655. [PMID: 37344566 PMCID: PMC10284893 DOI: 10.1038/s42003-023-05034-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 06/12/2023] [Indexed: 06/23/2023] Open
Abstract
Alzheimer's disease (AD) is characterized by brain plaques, tangles, and cognitive impairment. AD is one of the most common age-related dementias in humans. Progress in characterizing AD and other age-related disorders is hindered by a perceived dearth of animal models that naturally reproduce diseases observed in humans. Mice and nonhuman primates are model systems used to understand human diseases. Still, these model systems lack many of the biological characteristics of Alzheimer-like diseases (e.g., plaques, tangles) as they grow older. In contrast, companion animal models (cats and dogs) age in ways that resemble humans. Both companion animal models and humans show evidence of brain atrophy, plaques, and tangles, as well as cognitive decline with age. We embrace a One Health perspective, which recognizes that the health of humans is connected to those of animals, and we illustrate how such a perspective can work synergistically to enhance human and animal health. A comparative biology perspective is ideally suited to integrate insights across veterinary and human medical disciplines and solve long-standing problems in aging.
Collapse
Affiliation(s)
- Alexandra A de Sousa
- Centre for Health and Cognition, Bath Spa University, Bath, UK
- Department of Psychology, University of Bath, Bath, UK
| | - Brier A Rigby Dames
- Department of Psychology, University of Bath, Bath, UK
- Department of Computer Science, University of Bath, Bath, UK
- Department of Biology and Biochemistry, Milner Centre for Evolution, University of Bath, Bath, UK
| | - Emily C Graff
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Rania Mohamedelhassan
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Tatianna Vassilopoulos
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Christine J Charvet
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA.
| |
Collapse
|
28
|
Stanley MC, McNaughton EJ, Fewster RM, Galbraith JA. Cumulative propagule pressure exerted by escaped pet parrots. J Appl Ecol 2023. [DOI: 10.1111/1365-2664.14341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Margaret C. Stanley
- Centre for Biodiversity and Biosecurity, School of Biological Sciences Waipapa Taumata Rau/University of Auckland Auckland New Zealand
| | - Ellery J. McNaughton
- Centre for Biodiversity and Biosecurity, School of Biological Sciences Waipapa Taumata Rau/University of Auckland Auckland New Zealand
- Auckland Council Auckland New Zealand
| | - Rachel M. Fewster
- Department of Statistics Waipapa Taumata Rau/University of Auckland Auckland New Zealand
| | - Josie A. Galbraith
- Centre for Biodiversity and Biosecurity, School of Biological Sciences Waipapa Taumata Rau/University of Auckland Auckland New Zealand
- Department of Natural Sciences Auckland Museum Auckland New Zealand
| |
Collapse
|
29
|
Stevanovic M, Lazic A, Schwirtlich M, Stanisavljevic Ninkovic D. The Role of SOX Transcription Factors in Ageing and Age-Related Diseases. Int J Mol Sci 2023; 24:851. [PMID: 36614288 PMCID: PMC9821406 DOI: 10.3390/ijms24010851] [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: 11/30/2022] [Revised: 12/27/2022] [Accepted: 12/28/2022] [Indexed: 01/05/2023] Open
Abstract
The quest for eternal youth and immortality is as old as humankind. Ageing is an inevitable physiological process accompanied by many functional declines that are driving factors for age-related diseases. Stem cell exhaustion is one of the major hallmarks of ageing. The SOX transcription factors play well-known roles in self-renewal and differentiation of both embryonic and adult stem cells. As a consequence of ageing, the repertoire of adult stem cells present in various organs steadily declines, and their dysfunction/death could lead to reduced regenerative potential and development of age-related diseases. Thus, restoring the function of aged stem cells, inducing their regenerative potential, and slowing down the ageing process are critical for improving the health span and, consequently, the lifespan of humans. Reprograming factors, including SOX family members, emerge as crucial players in rejuvenation. This review focuses on the roles of SOX transcription factors in stem cell exhaustion and age-related diseases, including neurodegenerative diseases, visual deterioration, chronic obstructive pulmonary disease, osteoporosis, and age-related cancers. A better understanding of the molecular mechanisms of ageing and the roles of SOX transcription factors in this process could open new avenues for developing novel strategies that will delay ageing and prevent age-related diseases.
Collapse
Affiliation(s)
- Milena Stevanovic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia
- Faculty of Biology, University of Belgrade, Studentski trg 16, 11158 Belgrade, Serbia
- Serbian Academy of Sciences and Arts, Knez Mihailova 35, 11000 Belgrade, Serbia
| | - Andrijana Lazic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia
| | - Marija Schwirtlich
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia
| | | |
Collapse
|
30
|
Möller S, Saul N, Projahn E, Barrantes I, Gézsi A, Walter M, Antal P, Fuellen G. Gene co-expression analyses of health(span) across multiple species. NAR Genom Bioinform 2022; 4:lqac083. [PMID: 36458022 PMCID: PMC9706456 DOI: 10.1093/nargab/lqac083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 08/20/2022] [Accepted: 10/31/2022] [Indexed: 12/03/2022] Open
Abstract
Health(span)-related gene clusters/modules were recently identified based on knowledge about the cross-species genetic basis of health, to interpret transcriptomic datasets describing health-related interventions. However, the cross-species comparison of health-related observations reveals a lot of heterogeneity, not least due to widely varying health(span) definitions and study designs, posing a challenge for the exploration of conserved healthspan modules and, specifically, their transfer across species. To improve the identification and exploration of conserved/transferable healthspan modules, here we apply an established workflow based on gene co-expression network analyses employing GEO/ArrayExpress data for human and animal models, and perform a comprehensive meta-study of the resulting modules related to health(span), yielding a small set of literature backed health(span) candidate genes. For each experiment, WGCNA (weighted gene correlation network analysis) was used to infer modules of genes which correlate in their expression with a 'health phenotype score' and to determine the most-connected (hub) genes (and their interactions) for each such module. After mapping these hub genes to their human orthologs, 12 health(span) genes were identified in at least two species (ACTN3, ANK1, MRPL18, MYL1, PAXIP1, PPP1CA, SCN3B, SDCBP, SKIV2L, TUBG1, TYROBP, WIPF1), for which enrichment analysis by g:profiler found an association with actin filament-based movement and associated organelles, as well as muscular structures. We conclude that a meta-study of hub genes from co-expression network analyses for the complex phenotype health(span), across multiple species, can yield molecular-mechanistic insights and can direct experimentalists to further investigate the contribution of individual genes and their interactions to health(span).
Collapse
Affiliation(s)
- Steffen Möller
- To whom correspondence should be addressed. Tel: +49 381 494 7361; Fax: +49 381 494 7203;
| | - Nadine Saul
- Humboldt-University of Berlin, Institute of Biology, Berlin, Germany
| | - Elias Projahn
- Rostock University Medical Center, Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock, Germany
| | - Israel Barrantes
- Rostock University Medical Center, Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock, Germany
| | - András Gézsi
- Budapest University of Technology and Economics, Department of Measurement and Information Systems, Budapest, Hungary
| | - Michael Walter
- Rostock University Medical Center, Institute for Clinical Chemistry and Laboratory Medicine, Rostock, Germany
| | - Péter Antal
- Budapest University of Technology and Economics, Department of Measurement and Information Systems, Budapest, Hungary
| | - Georg Fuellen
- Rostock University Medical Center, Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock, Germany
| |
Collapse
|
31
|
Amson E, Scheyer TM, Martinez Q, Schwermann AH, Koyabu D, He K, Ziegler R. Unique bone microanatomy reveals ancestry of subterranean specializations in mammals. Evol Lett 2022; 6:552-561. [PMID: 36579164 PMCID: PMC9783445 DOI: 10.1002/evl3.303] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 09/06/2022] [Accepted: 09/30/2022] [Indexed: 11/13/2022] Open
Abstract
Acquiring a subterranean lifestyle entails a substantial shift for many aspects of terrestrial vertebrates' biology. Although this lifestyle is associated with multiple instances of convergent evolution, the relative success of some subterranean lineages largely remains unexplained. Here, we focus on the mammalian transitions to life underground, quantifying bone microanatomy through high-resolution X-ray tomography. The true moles stand out in this dataset. Examination of this family's bone histology reveals that the highly fossorial moles acquired a unique phenotype involving large amounts of compacted coarse cancellous bone. This phenotype exceeds the adaptive optimum seemingly shared by several other subterranean mammals and can be traced back to some of the first known members of the family. This remarkable microanatomy was acquired early in the history of the group and evolved faster than the gross morphology innovations of true moles' forelimb. This echoes the pattern described for other lifestyle transitions, such as the acquisition of bone mass specializations in secondarily aquatic tetrapods. Highly plastic traits-such as those pertaining to bone structure-are hence involved in the early stages of different types of lifestyle transitions.
Collapse
Affiliation(s)
- Eli Amson
- Staatliches Museum für Naturkunde StuttgartDE‐70191StuttgartGermany
| | - Torsten M. Scheyer
- Palaeontological Institute and MuseumUniversity of ZurichZurichCH‐8006Switzerland
| | - Quentin Martinez
- Staatliches Museum für Naturkunde StuttgartDE‐70191StuttgartGermany
| | - Achim H. Schwermann
- LWL‐Museum für NaturkundeWestfälisches Landesmuseum mit PlanetariumDE‐48161MünsterGermany
| | - Daisuke Koyabu
- Research and Development Center for Precision MedicineUniversity of TsukubaTsukuba305‐8550Japan
| | - Kai He
- Key Laboratory of Conservation and Application in Biodiversity of South China, School of Life SciencesGuangzhou UniversityGuangzhou510006China
| | - Reinhard Ziegler
- Staatliches Museum für Naturkunde StuttgartDE‐70191StuttgartGermany
| |
Collapse
|
32
|
Mikhailova AG, Mikhailova AA, Ushakova K, Tretiakov EO, Iliushchenko D, Shamansky V, Lobanova V, Kozenkov I, Efimenko B, Yurchenko AA, Kozenkova E, Zdobnov EM, Makeev V, Yurov V, Tanaka M, Gostimskaya I, Fleischmann Z, Annis S, Franco M, Wasko K, Denisov S, Kunz WS, Knorre D, Mazunin I, Nikolaev S, Fellay J, Reymond A, Khrapko K, Gunbin K, Popadin K. A mitochondria-specific mutational signature of aging: increased rate of A > G substitutions on the heavy strand. Nucleic Acids Res 2022; 50:10264-10277. [PMID: 36130228 PMCID: PMC9561281 DOI: 10.1093/nar/gkac779] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 08/02/2022] [Accepted: 09/07/2022] [Indexed: 11/21/2022] Open
Abstract
The mutational spectrum of the mitochondrial DNA (mtDNA) does not resemble any of the known mutational signatures of the nuclear genome and variation in mtDNA mutational spectra between different organisms is still incomprehensible. Since mitochondria are responsible for aerobic respiration, it is expected that mtDNA mutational spectrum is affected by oxidative damage. Assuming that oxidative damage increases with age, we analyse mtDNA mutagenesis of different species in regards to their generation length. Analysing, (i) dozens of thousands of somatic mtDNA mutations in samples of different ages (ii) 70053 polymorphic synonymous mtDNA substitutions reconstructed in 424 mammalian species with different generation lengths and (iii) synonymous nucleotide content of 650 complete mitochondrial genomes of mammalian species we observed that the frequency of AH > GH substitutions (H: heavy strand notation) is twice bigger in species with high versus low generation length making their mtDNA more AH poor and GH rich. Considering that AH > GH substitutions are also sensitive to the time spent single-stranded (TSSS) during asynchronous mtDNA replication we demonstrated that AH > GH substitution rate is a function of both species-specific generation length and position-specific TSSS. We propose that AH > GH is a mitochondria-specific signature of oxidative damage associated with both aging and TSSS.
Collapse
Affiliation(s)
- Alina G Mikhailova
- Center for Mitochondrial Functional Genomics, Immanuel Kant Baltic Federal University, Kaliningrad, Russian Federation
- Vavilov Institute of General Genetics RAS, Moscow, Russia
| | - Alina A Mikhailova
- Center for Mitochondrial Functional Genomics, Immanuel Kant Baltic Federal University, Kaliningrad, Russian Federation
| | - Kristina Ushakova
- Center for Mitochondrial Functional Genomics, Immanuel Kant Baltic Federal University, Kaliningrad, Russian Federation
| | - Evgeny O Tretiakov
- Center for Mitochondrial Functional Genomics, Immanuel Kant Baltic Federal University, Kaliningrad, Russian Federation
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Dmitrii Iliushchenko
- Center for Mitochondrial Functional Genomics, Immanuel Kant Baltic Federal University, Kaliningrad, Russian Federation
| | - Victor Shamansky
- Center for Mitochondrial Functional Genomics, Immanuel Kant Baltic Federal University, Kaliningrad, Russian Federation
| | - Valeria Lobanova
- Center for Mitochondrial Functional Genomics, Immanuel Kant Baltic Federal University, Kaliningrad, Russian Federation
| | - Ivan Kozenkov
- Center for Mitochondrial Functional Genomics, Immanuel Kant Baltic Federal University, Kaliningrad, Russian Federation
| | - Bogdan Efimenko
- Center for Mitochondrial Functional Genomics, Immanuel Kant Baltic Federal University, Kaliningrad, Russian Federation
| | - Andrey A Yurchenko
- INSERM U981, Gustave Roussy Cancer Campus, Université Paris Saclay, Villejuif, France
| | - Elena Kozenkova
- Institute of Physics, Mathematics and Information Technology, Immanuel Kant Baltic Federal University, Kaliningrad, Russian Federation
| | - Evgeny M Zdobnov
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Vsevolod Makeev
- Vavilov Institute of General Genetics RAS, Moscow, Russia
- Moscow Institute of Physics and Technology, Moscow, Russian Federation
| | - Valerian Yurov
- Institute of Physics, Mathematics and Information Technology, Immanuel Kant Baltic Federal University, Kaliningrad, Russian Federation
| | - Masashi Tanaka
- Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Irina Gostimskaya
- Manchester Institute of Biotechnology, The University of Manchester, Manchester, United Kingdom
| | - Zoe Fleischmann
- Department of Biology, Northeastern University, Boston, MA, USA
| | - Sofia Annis
- Department of Biology, Northeastern University, Boston, MA, USA
| | - Melissa Franco
- Department of Biology, Northeastern University, Boston, MA, USA
| | - Kevin Wasko
- Department of Biology, Northeastern University, Boston, MA, USA
| | - Stepan Denisov
- Center for Mitochondrial Functional Genomics, Immanuel Kant Baltic Federal University, Kaliningrad, Russian Federation
- School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Wolfram S Kunz
- Department of Epileptology and Institute of Experimental Epileptology and Cognition Research, University Bonn, Bonn, Germany
| | - Dmitry Knorre
- The A.N. Belozersky Institute Of Physico-Chemical Biology, Moscow State University, Moscow, Russian Federation
| | - Ilya Mazunin
- Center for Molecular and Cellular Biology, Skolkovo Institute of Science and Technology (Skoltech), Skolkovo, Russian Federation
- Fomin Clinic, Moscow, Russian Federation
- Medical Genomics LLC, Moscow, Russian Federation
| | - Sergey Nikolaev
- INSERM U981, Gustave Roussy Cancer Campus, Université Paris Saclay, Villejuif, France
| | - Jacques Fellay
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Alexandre Reymond
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | | | - Konstantin Gunbin
- Center for Mitochondrial Functional Genomics, Immanuel Kant Baltic Federal University, Kaliningrad, Russian Federation
- Institute of Molecular and Cellular Biology SB RAS, Novosibirsk, Russian Federation
| | - Konstantin Popadin
- Center for Mitochondrial Functional Genomics, Immanuel Kant Baltic Federal University, Kaliningrad, Russian Federation
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
33
|
Teeth, prenatal growth rates, and the evolution of human-like pregnancy in later Homo. Proc Natl Acad Sci U S A 2022; 119:e2200689119. [PMID: 36191229 PMCID: PMC9564099 DOI: 10.1073/pnas.2200689119] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Evidence of how gestational parameters evolved is essential to understanding this fundamental stage of human life. Until now, these data seemed elusive given the skeletal bias of the fossil record. We demonstrate that dentition provides a window into the life of neonates. Teeth begin to form in utero and are intimately associated with gestational development. We measured the molar dentition for 608 catarrhine primates and collected data on prenatal growth rate (PGR) and endocranial volume (ECV) for 19 primate genera from the literature. We found that PGR and ECV are highly correlated (R2 = 0.93, P < 0.001). Additionally, we demonstrated that molar proportions are significantly correlated with PGR (P = 0.004) and log-transformed ECV (P = 0.001). From these correlations, we developed two methods for reconstructing PGR in the fossil record, one using ECV and one using molar proportions. Dental proportions reconstruct hominid ECV (R2 = 0.81, P < 0.001), a result that can be extrapolated to PGR. As teeth dominate fossil assemblages, our findings greatly expand our ability to investigate life history in the fossil record. Fossil ECVs and dental measurements from 13 hominid species both support significantly increasing PGR throughout the terminal Miocene and Plio-Pleistocene, reflecting known evolutionary changes. Together with pelvic and endocranial morphology, reconstructed PGRs indicate the need for increasing maternal energetics during pregnancy over the last 6 million years, reaching a human-like PGR (i.e., more similar to humans than to other extant apes) and ECV in later Homo less than 1 million years ago.
Collapse
|
34
|
Therapeutic Antiaging Strategies. Biomedicines 2022; 10:biomedicines10102515. [PMID: 36289777 PMCID: PMC9599338 DOI: 10.3390/biomedicines10102515] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/21/2022] [Accepted: 09/24/2022] [Indexed: 11/17/2022] Open
Abstract
Aging constitutes progressive physiological changes in an organism. These changes alter the normal biological functions, such as the ability to manage metabolic stress, and eventually lead to cellular senescence. The process itself is characterized by nine hallmarks: genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, deregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, and altered intercellular communication. These hallmarks are risk factors for pathologies, such as cardiovascular diseases, neurodegenerative diseases, and cancer. Emerging evidence has been focused on examining the genetic pathways and biological processes in organisms surrounding these nine hallmarks. From here, the therapeutic approaches can be addressed in hopes of slowing the progression of aging. In this review, data have been collected on the hallmarks and their relative contributions to aging and supplemented with in vitro and in vivo antiaging research experiments. It is the intention of this article to highlight the most important antiaging strategies that researchers have proposed, including preventive measures, systemic therapeutic agents, and invasive procedures, that will promote healthy aging and increase human life expectancy with decreased side effects.
Collapse
|
35
|
Piloto JH, Rodriguez M, Choe KP. Sexual dimorphism in Caenorhabditis elegans stress resistance. PLoS One 2022; 17:e0272452. [PMID: 35951614 PMCID: PMC9371273 DOI: 10.1371/journal.pone.0272452] [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: 12/01/2021] [Accepted: 07/19/2022] [Indexed: 11/18/2022] Open
Abstract
Physiological responses to the environment, disease, and aging vary by sex in many animals, but mechanisms of dimorphism have only recently begun to receive careful attention. The genetic model nematode Caenorhabditis elegans has well-defined mechanisms of stress response, aging, and sexual differentiation. C. elegans has males, but the vast majority of research only uses hermaphrodites. We found that males of the standard N2 laboratory strain were more resistant to hyperosmolarity, heat, and a natural pro-oxidant than hermaphrodites when in mixed-sex groups. Resistance to heat and pro-oxidant were also male-biased in three genetically and geographically diverse C. elegans strains consistent with a species-wide dimorphism that is not specific to domestication. N2 males were also more resistant to heat and pro-oxidant when keep individually indicating that differences in resistance do not require interactions between worms. We found that males induce canonical stress response genes by similar degrees and in similar tissues as hermaphrodites suggesting the importance of other mechanisms. We find that resistance to heat and pro-oxidant are influenced by the sex differentiation transcription factor TRA-1 suggesting that downstream organ differentiation pathways establish differences in stress resistance. Environmental stress influences survival in natural environments, degenerative disease, and aging. Understanding mechanisms of stress response dimorphism can therefore provide insights into sex-specific population dynamics, disease, and longevity.
Collapse
Affiliation(s)
- Juan H. Piloto
- Department of Biology and Genetics Institute, University of Florida, Gainesville, FL, United States of America
| | - Michael Rodriguez
- Department of Biology and Genetics Institute, University of Florida, Gainesville, FL, United States of America
| | - Keith P. Choe
- Department of Biology and Genetics Institute, University of Florida, Gainesville, FL, United States of America
- * E-mail:
| |
Collapse
|
36
|
Cohen AA, Ferrucci L, Fülöp T, Gravel D, Hao N, Kriete A, Levine ME, Lipsitz LA, Olde Rikkert MGM, Rutenberg A, Stroustrup N, Varadhan R. A complex systems approach to aging biology. NATURE AGING 2022; 2:580-591. [PMID: 37117782 DOI: 10.1038/s43587-022-00252-6] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 06/08/2022] [Indexed: 04/30/2023]
Abstract
Having made substantial progress understanding molecules, cells, genes and pathways, aging biology research is now moving toward integration of these parts, attempting to understand how their joint dynamics may contribute to aging. Such a shift of perspective requires the adoption of a formal complex systems framework, a transition being facilitated by large-scale data collection and new analytical tools. Here, we provide a theoretical framework to orient researchers around key concepts for this transition, notably emergence, interaction networks and resilience. Drawing on evolutionary theory, network theory and principles of homeostasis, we propose that organismal function is accomplished by the integration of regulatory mechanisms at multiple hierarchical scales, and that the disruption of this ensemble causes the phenotypic and functional manifestations of aging. We present key examples at scales ranging from sub-organismal biology to clinical geriatrics, outlining how this approach can potentially enrich our understanding of aging.
Collapse
Affiliation(s)
- Alan A Cohen
- PRIMUS Research Group, Department of Family Medicine, University of Sherbrooke, Sherbrooke, Quebec, Canada.
- Research Center on Aging and Research Center of Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Quebec, Canada.
- Butler Columbia Aging Center and Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA.
| | - Luigi Ferrucci
- Intramural Research Program of the National Institute on Aging, Baltimore, MD, USA
| | - Tamàs Fülöp
- Research Center on Aging and Research Center of Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Quebec, Canada
- Department of Medicine, Geriatric Division, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Dominique Gravel
- Department of Biology, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Nan Hao
- Section of Molecular Biology, Division of Biological Sciences, University of California, San Diego, San Diego, CA, USA
| | - Andres Kriete
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Morgan E Levine
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Lewis A Lipsitz
- Beth Israel Deaconess Medical Center, Hebrew SeniorLife, Hinda and Arthur Marcus Institute for Aging Research, and Harvard Medical School, Boston, MA, USA
| | | | - Andrew Rutenberg
- Department of Physics and Atmospheric Science, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Nicholas Stroustrup
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Ravi Varadhan
- Department of Oncology, Quantitative Sciences Division, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
37
|
Hsu B, Pakanen V, Boner W, Doligez B, Eeva T, Groothuis TGG, Korpimäki E, Laaksonen T, Lelono A, Monaghan P, Sarraude T, Thomson RL, Tolvanen J, Tschirren B, Vásquez RA, Ruuskanen S. Maternally transferred thyroid hormones and life-history variation in birds. J Anim Ecol 2022; 91:1489-1506. [PMID: 35470435 PMCID: PMC9546341 DOI: 10.1111/1365-2656.13708] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 03/21/2022] [Indexed: 12/04/2022]
Abstract
In vertebrates, thyroid hormones (THs) play an important role in the regulation of growth, development, metabolism, photoperiodic responses and migration. Maternally transferred THs are important for normal early phase embryonic development when embryos are not able to produce endogenous THs. Previous studies have shown that variation in maternal THs within the physiological range can influence offspring phenotype. Given the essential functions of maternal THs in development and metabolism, THs may be a mediator of life-history variation across species. We tested the hypothesis that differences in life histories are associated with differences in maternal TH transfer across species. Using birds as a model, we specifically tested whether maternally transferred yolk THs covary with migratory status, developmental mode and traits related to pace-of-life (e.g. basal metabolic rate, maximum life span). We collected un-incubated eggs (n = 1-21 eggs per species, median = 7) from 34 wild and captive bird species across 17 families and six orders to measure yolk THs [both triiodothyronine (T3) and thyroxine (T4)], compiled life-history trait data from the literature and used Bayesian phylogenetic mixed models to test our hypotheses. Our models indicated that both concentrations and total amounts of the two main forms of THs (T3 and T4) were higher in the eggs of migratory species compared to resident species, and total amounts were higher in the eggs of precocial species, which have longer prenatal developmental periods, than in those of altricial species. However, maternal yolk THs did not show clear associations with pace-of-life-related traits, such as fecundity, basal metabolic rate or maximum life span. We quantified interspecific variation in maternal yolk THs in birds, and our findings suggest higher maternal TH transfer is associated with the precocial mode of development and migratory status. Whether maternal THs represent a part of the mechanism underlying the evolution of precocial development and migration or a consequence of such life histories is currently unclear. We therefore encourage further studies to explore the physiological mechanisms and evolutionary processes underlying these patterns.
Collapse
Affiliation(s)
- Bin‐Yan Hsu
- Department of BiologyUniversity of TurkuTurkuFinland
| | - Veli‐Matti Pakanen
- Ecology and Genetics Research UnitUniversity of OuluOuluFinland
- Department of Biological and Environmental SciencesUniversity of GothenburgGothenburgSweden
| | - Winnie Boner
- Institute of Biodiversity, Animal Healthy and Comparative MedicineUniversity of GlasgowGlasgowUK
| | - Blandine Doligez
- Department of Biometry and Evolutionary Biology, CNRS UMR 5558Université de Lyon 1VilleurbanneFrance
| | - Tapio Eeva
- Department of BiologyUniversity of TurkuTurkuFinland
| | - Ton G. G. Groothuis
- Groningen Institute for Evolutionary Life Sciences (GELIFES)University of GroningenGroningenThe Netherlands
| | | | | | - Asmoro Lelono
- Groningen Institute for Evolutionary Life Sciences (GELIFES)University of GroningenGroningenThe Netherlands
- Biology Department, Natural Sciences and Mathematics FacultyJember University of IndonesiaJemberIndonesia
| | - Pat Monaghan
- Institute of Biodiversity, Animal Healthy and Comparative MedicineUniversity of GlasgowGlasgowUK
| | - Tom Sarraude
- Department of BiologyUniversity of TurkuTurkuFinland
- Groningen Institute for Evolutionary Life Sciences (GELIFES)University of GroningenGroningenThe Netherlands
| | - Robert L. Thomson
- Fitzpatrick Institute of African Ornithology, DST‐NRF Centre of ExcellenceUniversity of Cape TownCape TownSouth Africa
| | - Jere Tolvanen
- Ecology and Genetics Research UnitUniversity of OuluOuluFinland
| | | | - Rodrigo A. Vásquez
- Instituto de Ecología y Biodiversidad, Departamento de Ciencias Ecológicas, Facultad de CienciasUniversidad de ChileSantiagoChile
| | - Suvi Ruuskanen
- Department of BiologyUniversity of TurkuTurkuFinland
- Department of Biological and Environmental SciencesUniversity of JyväskyläJyväskyläFinland
| |
Collapse
|
38
|
Jackson J, Le Coeur C, Jones O. Life-history predicts global population responses to the weather in terrestrial mammals. eLife 2022; 11:74161. [PMID: 35775734 PMCID: PMC9307275 DOI: 10.7554/elife.74161] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 06/30/2022] [Indexed: 11/26/2022] Open
Abstract
With the looming threat of abrupt ecological disruption due to a changing climate, predicting which species are most vulnerable to environmental change is critical. The life-history of a species is an evolved response to its environmental context, and therefore a promising candidate for explaining differences in climate-change responses. However, we need broad empirical assessments from across the world's ecosystems to explore the link between life history and climate-change responses. Here, we use long-term abundance records from 157 species of terrestrial mammals and a two-step Bayesian meta-regression framework to investigate the link between annual weather anomalies, population growth rates, and species-level life history. Overall, we found no directional effect of temperature or precipitation anomalies or variance on annual population growth rates. Furthermore, population responses to weather anomalies were not predicted by phylogenetic covariance, and instead there was more variability in weather responses for populations within a species. Crucially, however, long-lived mammals with smaller litter sizes had smaller absolute population responses to weather anomalies compared with their shorter living counterparts with larger litters. These results highlight the role of species-level life history in driving responses to the environment.
Collapse
Affiliation(s)
- John Jackson
- 2.Department of Zoology, University of Oxford, Oxford, United Kingdom
| | | | - Owen Jones
- Department of Biology, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
39
|
Li Q, Milenkovic T. Supervised Prediction of Aging-Related Genes From a Context-Specific Protein Interaction Subnetwork. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2022; 19:2484-2498. [PMID: 33929964 DOI: 10.1109/tcbb.2021.3076961] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Human aging is linked to many prevalent diseases. The aging process is highly influenced by genetic factors. Hence, it is important to identify human aging-related genes. We focus on supervised prediction of such genes. Gene expression-based methods for this purpose study genes in isolation from each other. While protein-protein interaction (PPI) network-based methods for this purpose account for interactions between genes' protein products, current PPI network data are context-unspecific, spanning different biological conditions. Instead, here, we focus on an aging-specific subnetwork of the entire PPI network, obtained by integrating aging-specific gene expression data and PPI network data. The potential of such data integration has been recognized but mostly in the context of cancer. So, we are the first to propose a supervised learning framework for predicting aging-related genes from an aging-specific PPI subnetwork. In a systematic and comprehensive evaluation, we find that in many of the evaluation tests: (i) using an aging-specific subnetwork indeed yields more accurate aging-related gene predictions than using the entire network, and (ii) predictive methods from our framework that have not previously been used for supervised prediction of aging-related genes outperform existing prominent methods for the same purpose. These results justify the need for our framework.
Collapse
|
40
|
Lu JY, Simon M, Zhao Y, Ablaeva J, Corson N, Choi Y, Yamada KYH, Schork NJ, Hood WR, Hill GE, Miller RA, Seluanov A, Gorbunova V. Comparative transcriptomics reveals circadian and pluripotency networks as two pillars of longevity regulation. Cell Metab 2022; 34:836-856.e5. [PMID: 35580607 PMCID: PMC9364679 DOI: 10.1016/j.cmet.2022.04.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/24/2022] [Accepted: 04/22/2022] [Indexed: 01/24/2023]
Abstract
Mammals differ more than 100-fold in maximum lifespan. Here, we conducted comparative transcriptomics on 26 species with diverse lifespans. We identified thousands of genes with expression levels negatively or positively correlated with a species' maximum lifespan (Neg- or Pos-MLS genes). Neg-MLS genes are primarily involved in energy metabolism and inflammation. Pos-MLS genes show enrichment in DNA repair, microtubule organization, and RNA transport. Expression of Neg- and Pos-MLS genes is modulated by interventions, including mTOR and PI3K inhibition. Regulatory networks analysis showed that Neg-MLS genes are under circadian regulation possibly to avoid persistent high expression, whereas Pos-MLS genes are targets of master pluripotency regulators OCT4 and NANOG and are upregulated during somatic cell reprogramming. Pos-MLS genes are highly expressed during embryogenesis but significantly downregulated after birth. This work provides targets for anti-aging interventions by defining pathways correlating with longevity across mammals and uncovering circadian and pluripotency networks as central regulators of longevity.
Collapse
Affiliation(s)
- J Yuyang Lu
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
| | - Matthew Simon
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
| | - Yang Zhao
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
| | - Julia Ablaeva
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
| | - Nancy Corson
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
| | - Yongwook Choi
- Quantitative Medicine and Systems Biology Division, Translational Genomics Research Institute, Phoenix, AZ 85004, USA
| | - KayLene Y H Yamada
- Department of Biological Sciences, Auburn University, Auburn, AL 36849, USA
| | - Nicholas J Schork
- Quantitative Medicine and Systems Biology Division, Translational Genomics Research Institute, Phoenix, AZ 85004, USA
| | - Wendy R Hood
- Department of Biological Sciences, Auburn University, Auburn, AL 36849, USA
| | - Geoffrey E Hill
- Department of Biological Sciences, Auburn University, Auburn, AL 36849, USA
| | - Richard A Miller
- Department of Pathology and Geriatrics Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Andrei Seluanov
- Department of Biology, University of Rochester, Rochester, NY 14627, USA.
| | - Vera Gorbunova
- Department of Biology, University of Rochester, Rochester, NY 14627, USA.
| |
Collapse
|
41
|
Baker P, Cooper-Mullin CM, Jimenez AG. Differences in advanced glycation endproducts (AGEs) in plasma from birds and mammals of different body sizes and ages. Comp Biochem Physiol A Mol Integr Physiol 2022; 267:111164. [PMID: 35158049 DOI: 10.1016/j.cbpa.2022.111164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/23/2022] [Accepted: 02/08/2022] [Indexed: 11/24/2022]
Abstract
Birds and mammals provide a physiological paradox: similar-sized mammals live shorter lives than birds; yet, birds have higher blood glucose concentrations than mammals, and higher basal metabolic rates. We have previously shown that oxidative stress patterns between mammals and birds differ, so that birds, generally, have lower blood antioxidant capacity, and lower lipid peroxidation concentration. There is a close association between oxidative stress and the production of carbohydrate-based damaged biomolecules, Advanced Glycation End-products (AGEs). In mammals, AGEs can bind to their receptor (RAGE), which can lead to increases in reactive oxygen species (ROS) production, and can decrease antioxidant capacity. Here, we used plasma from birds and mammals to address whether blood plasma AGE-BSA concentration is associated with body mass and age in these two groups. We found a statistically significantly higher average concentrations of AGE-BSA in birds compared with mammals, and we found a significantly positive correlation between AGE-BSA and age in mammals, though, this correlation disappeared after phylogenetic correction. We propose that the higher AGE concentration in birds is mainly attributable to greater AGE-production due to elevated basal glucose concentrations and decreased AGE-clearance given differences in glomerular filtration rates in birds compared with mammals. Additionally, due to the potential lack of an AGE receptor in birds, AGE accumulation may not be closely linked to oxidative stress and therefore pose a lesser physiological challenge in birds compared to mammals.
Collapse
Affiliation(s)
- Peter Baker
- Colgate University, Department of Biology, 13 Oak Dr., Hamilton, NY 13346, United States of America
| | - Clara M Cooper-Mullin
- University of Rhode Island, Natural Resources Science, 1 Greenhouse Drive, Kingston, RI 02881, United States of America
| | - Ana Gabriela Jimenez
- Colgate University, Department of Biology, 13 Oak Dr., Hamilton, NY 13346, United States of America.
| |
Collapse
|
42
|
Hiratani N, Latham PE. Developmental and evolutionary constraints on olfactory circuit selection. Proc Natl Acad Sci U S A 2022; 119:e2100600119. [PMID: 35263217 PMCID: PMC8931209 DOI: 10.1073/pnas.2100600119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 01/14/2022] [Indexed: 11/18/2022] Open
Abstract
SignificanceIn this work, we explore the hypothesis that biological neural networks optimize their architecture, through evolution, for learning. We study early olfactory circuits of mammals and insects, which have relatively similar structure but a huge diversity in size. We approximate these circuits as three-layer networks and estimate, analytically, the scaling of the optimal hidden-layer size with input-layer size. We find that both longevity and information in the genome constrain the hidden-layer size, so a range of allometric scalings is possible. However, the experimentally observed allometric scalings in mammals and insects are consistent with biologically plausible values. This analysis should pave the way for a deeper understanding of both biological and artificial networks.
Collapse
Affiliation(s)
- Naoki Hiratani
- Gatsby Computational Neuroscience Unit, University College London, London W1T 4JG, United Kingdom
| | - Peter E. Latham
- Gatsby Computational Neuroscience Unit, University College London, London W1T 4JG, United Kingdom
| |
Collapse
|
43
|
Oliviero G, Kovalchuk S, Rogowska-Wrzesinska A, Schwämmle V, Jensen ON. Distinct and diverse chromatin-proteomes of ageing mouse organs reveal protein signatures that correlate with physiological functions. eLife 2022; 11:73524. [PMID: 35259090 PMCID: PMC8933006 DOI: 10.7554/elife.73524] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
Temporal molecular changes in ageing mammalian organs are of relevance to disease aetiology because many age-related diseases are linked to changes in the transcriptional and epigenetic machinery that regulate gene expression. We performed quantitative proteome analysis of chromatin-enriched protein extracts to investigate the dynamics of the chromatin proteomes of the mouse brain, heart, lung, kidney, liver, and spleen at 3, 5, 10, and 15 months of age. Each organ exhibited a distinct chromatin proteome and sets of unique proteins. The brain and spleen chromatin proteomes were the most extensive, diverse, and heterogenous among the six organs. The spleen chromatin proteome appeared static during the lifespan, presenting a young phenotype that reflects the permanent alertness state and important role of this organ in physiological defence and immunity. We identified a total of 5928 proteins, including 2472 nuclear or chromatin-associated proteins across the six mouse organs. Up to 3125 proteins were quantified in each organ, demonstrating distinct and organ-specific temporal protein expression timelines and regulation at the post-translational level. Bioinformatics meta-analysis of these chromatin proteomes revealed distinct physiological and ageing-related features for each organ. Our results demonstrate the efficiency of organelle-specific proteomics for in vivo studies of a model organism and consolidate the hypothesis that chromatin-associated proteins are involved in distinct and specific physiological functions in ageing organs.
Collapse
Affiliation(s)
- Giorgio Oliviero
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Sergey Kovalchuk
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | | | - Veit Schwämmle
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Ole N Jensen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
44
|
Gübert J, Hahn‐Klimroth M, Dierkes PW. BOVIDS: A deep learning-based software package for pose estimation to evaluate nightly behavior and its application to common elands ( Tragelaphus oryx) in zoos. Ecol Evol 2022; 12:e8701. [PMID: 35342615 PMCID: PMC8928879 DOI: 10.1002/ece3.8701] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 02/13/2022] [Accepted: 02/17/2022] [Indexed: 12/29/2022] Open
Abstract
Only a few studies on the nocturnal behavior of African ungulates exist so far, with mostly small sample sizes. For a comprehensive understanding of nocturnal behavior, the data basis needs to be expanded. Results obtained by observing zoo animals can provide clues for the study of wild animals and furthermore contribute to a better understanding of animal welfare and better husbandry conditions in zoos. The current contribution reduces the lack of data in two ways. First, we present a stand-alone open-source software package based on deep learning techniques, named Behavioral Observations by Videos and Images using Deep-Learning Software (BOVIDS). It can be used to identify ungulates in their enclosure and to determine the three behavioral poses "Standing," "Lying-head up," and "Lying-head down" on 11,411 h of video material with an accuracy of 99.4%. Second, BOVIDS is used to conduct a case study on 25 common elands (Tragelaphus oryx) out of 5 EAZA zoos with a total of 822 nights, yielding the first detailed description of the nightly behavior of common elands. Our results indicate that age and sex are influencing factors on the nocturnal activity budget, the length of behavioral phases as well as the number of phases per behavioral state during the night while the keeping zoo has no significant influence. It is found that males spend more time in REM sleep posture than females while young animals spend more time in this position than adult ones. Finally, the results suggest a rhythm between the Standing and Lying phases among common elands that opens future research directions.
Collapse
Affiliation(s)
- Jennifer Gübert
- Faculty of Biological SciencesBioscience Education and Zoo BiologyGoethe UniversityFrankfurtGermany
| | | | - Paul W. Dierkes
- Faculty of Biological SciencesBioscience Education and Zoo BiologyGoethe UniversityFrankfurtGermany
| |
Collapse
|
45
|
Kim CK, Sachdev PS, Braidy N. Recent Neurotherapeutic Strategies to Promote Healthy Brain Aging: Are we there yet? Aging Dis 2022; 13:175-214. [PMID: 35111369 PMCID: PMC8782556 DOI: 10.14336/ad.2021.0705] [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: 04/14/2021] [Accepted: 07/05/2021] [Indexed: 12/21/2022] Open
Abstract
Owing to the global exponential increase in population ageing, there is an urgent unmet need to develop reliable strategies to slow down and delay the ageing process. Age-related neurodegenerative diseases are among the main causes of morbidity and mortality in our contemporary society and represent a major socio-economic burden. There are several controversial factors that are thought to play a causal role in brain ageing which are continuously being examined in experimental models. Among them are oxidative stress and brain inflammation which are empirical to brain ageing. Although some candidate drugs have been developed which reduce the ageing phenotype, their clinical translation is limited. There are several strategies currently in development to improve brain ageing. These include strategies such as caloric restriction, ketogenic diet, promotion of cellular nicotinamide adenine dinucleotide (NAD+) levels, removal of senescent cells, 'young blood' transfusions, enhancement of adult neurogenesis, stem cell therapy, vascular risk reduction, and non-pharmacological lifestyle strategies. Several studies have shown that these strategies can not only improve brain ageing by attenuating age-related neurodegenerative disease mechanisms, but also maintain cognitive function in a variety of pre-clinical experimental murine models. However, clinical evidence is limited and many of these strategies are awaiting findings from large-scale clinical trials which are nascent in the current literature. Further studies are needed to determine their long-term efficacy and lack of adverse effects in various tissues and organs to gain a greater understanding of their potential beneficial effects on brain ageing and health span in humans.
Collapse
Affiliation(s)
- Chul-Kyu Kim
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia
| | - Perminder S Sachdev
- Neuropsychiatric Institute, Euroa Centre, Prince of Wales Hospital, Sydney, Australia
| | - Nady Braidy
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia
| |
Collapse
|
46
|
Ramírez-Delgado JP, Di Marco M, Watson JEM, Johnson CJ, Rondinini C, Corredor Llano X, Arias M, Venter O. Matrix condition mediates the effects of habitat fragmentation on species extinction risk. Nat Commun 2022; 13:595. [PMID: 35105881 PMCID: PMC8807630 DOI: 10.1038/s41467-022-28270-3] [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: 04/07/2021] [Accepted: 01/18/2022] [Indexed: 11/10/2022] Open
Abstract
Habitat loss is the leading cause of the global decline in biodiversity, but the influence of human pressure within the matrix surrounding habitat fragments remains poorly understood. Here, we measure the relationship between fragmentation (the degree of fragmentation and the degree of patch isolation), matrix condition (measured as the extent of high human footprint levels), and the change in extinction risk of 4,426 terrestrial mammals. We find that the degree of fragmentation is strongly associated with changes in extinction risk, with higher predictive importance than life-history traits and human pressure variables. Importantly, we discover that fragmentation and the matrix condition are stronger predictors of risk than habitat loss and habitat amount. Moreover, the importance of fragmentation increases with an increasing deterioration of the matrix condition. These findings suggest that restoration of the habitat matrix may be an important conservation action for mitigating the negative effects of fragmentation on biodiversity. The influence of human pressure within the matrix surrounding habitat fragments remains poorly understood. This study measures the relationship between habitat fragmentation, matrix condition and the change in extinction risk of 4,426 terrestrial mammals, finding that fragmentation and matrix condition are stronger predictors of risk than habitat loss and habitat amount.
Collapse
Affiliation(s)
- Juan Pablo Ramírez-Delgado
- Natural Resources and Environmental Studies Institute, University of Northern British Columbia, Prince George, V2N 4Z9, Canada.
| | - Moreno Di Marco
- Department of Biology and Biotechnologies, Sapienza University of Rome, 00185, Rome, Italy
| | - James E M Watson
- School of Earth and Environmental Sciences, University of Queensland, St Lucia, 4072, Australia.,Centre for Biodiversity and Conservation Science, School of Biological Sciences, The University of Queensland, Brisbane, 4072, QLD, Australia
| | - Chris J Johnson
- Natural Resources and Environmental Studies Institute, University of Northern British Columbia, Prince George, V2N 4Z9, Canada
| | - Carlo Rondinini
- Global Mammal Assessment Program, Department of Biology and Biotechnologies, Sapienza University of Rome, Rome, 00185, Italy
| | - Xavier Corredor Llano
- Natural Resources and Environmental Studies Institute, University of Northern British Columbia, Prince George, V2N 4Z9, Canada
| | - Miguel Arias
- Natural Resources and Environmental Studies Institute, University of Northern British Columbia, Prince George, V2N 4Z9, Canada
| | - Oscar Venter
- Natural Resources and Environmental Studies Institute, University of Northern British Columbia, Prince George, V2N 4Z9, Canada
| |
Collapse
|
47
|
Zhao Y, Seluanov A, Gorbunova V. Revelations About Aging and Disease from Unconventional Vertebrate Model Organisms. Annu Rev Genet 2021; 55:135-159. [PMID: 34416119 PMCID: PMC8903061 DOI: 10.1146/annurev-genet-071719-021009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Aging is a major risk factor for multiple diseases. Understanding the underlying mechanisms of aging would help to delay and prevent age-associated diseases. Short-lived model organisms have been extensively used to study the mechanisms of aging. However, these short-lived species may be missing the longevity mechanisms that are needed to extend the lifespan of an already long-lived species such as humans. Unconventional long-lived animal species are an excellent resource to uncover novel mechanisms of longevity and disease resistance. Here, we review mechanisms that evolved in nonmodel vertebrate species to counteract age-associated diseases. Some antiaging mechanisms are conserved across species; however, various nonmodel species also evolved unique mechanisms to delay aging and prevent disease. This variety of antiaging mechanisms has evolved due to the remarkably diverse habitats and behaviors of these species. We propose that exploring a wider range of unconventional vertebrates will provide important resources to study antiaging mechanisms that are potentially applicable to humans.
Collapse
Affiliation(s)
- Yang Zhao
- Department of Biology, University of Rochester, Rochester, New York 14627, USA; ,
| | - Andrei Seluanov
- Department of Biology, University of Rochester, Rochester, New York 14627, USA; ,
| | - Vera Gorbunova
- Department of Biology, University of Rochester, Rochester, New York 14627, USA; ,
| |
Collapse
|
48
|
Glutamine Homeostasis and Its Role in the Adaptive Strategies of the Blind Mole Rat, Spalax. Metabolites 2021; 11:metabo11110755. [PMID: 34822413 PMCID: PMC8620300 DOI: 10.3390/metabo11110755] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 10/26/2021] [Accepted: 10/30/2021] [Indexed: 12/20/2022] Open
Abstract
Oxidative metabolism is fine-tuned machinery that combines two tightly coupled fluxes of glucose and glutamine-derived carbons. Hypoxia interrupts the coordination between the metabolism of these two nutrients and leads to a decrease of the system efficacy and may eventually cause cell death. The subterranean blind mole rat, Spalax, is an underexplored, underground, hypoxia-tolerant mammalian group which spends its life under sharply fluctuating oxygen levels. Primary Spalax cells are an exceptional model to study the metabolic strategies that have evolved in mammals inhabiting low-oxygen niches. In this study we explored the metabolic frame of glutamine (Gln) homeostasis in Spalax skin cells under normoxic and hypoxic conditions and their impacts on the metabolism of rat cells. Targeted metabolomics employing liquid chromatography and mass spectrometry (LC-MS) was used to track the fate of heavy glutamine carbons (13C5 Gln) after 24 h under normoxia or hypoxia (1% O2). Our results indicated that large amounts of glutamine-originated carbons were detected as proline (Pro) and hydroxyproline (HPro) in normoxic Spalax cells with a further increase under hypoxia, suggesting a strategy for reduced Gln carbons storage in proteins. The intensity of the flux and the presence of HPro suggests collagen as a candidate protein that is most abundant in animals, and as the primary source of HPro. An increased conversion of αKG to 2 HG that was indicated in hypoxic Spalax cells prevents the degradation of hypoxia-inducible factor 1α (HIF-1α) and, consequently, maintains cytosolic and mitochondrial carbons fluxes that were uncoupled via inhibition of the pyruvate dehydrogenase complex. A strong antioxidant defense in Spalax cells can be attributed, at least in part, to the massive usage of glutamine-derived glutamate for glutathione (GSH) production. The present study uncovers additional strategies that have evolved in this unique mammal to support its hypoxia tolerance, and probably contribute to its cancer resistance, longevity, and healthy aging.
Collapse
|
49
|
Miskevich D, Chaban A, Dronina M, Abramovich I, Gottlieb E, Shams I. Comprehensive Analysis of 13C 6 Glucose Fate in the Hypoxia-Tolerant Blind Mole Rat Skin Fibroblasts. Metabolites 2021; 11:metabo11110734. [PMID: 34822392 PMCID: PMC8621580 DOI: 10.3390/metabo11110734] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 10/17/2021] [Accepted: 10/25/2021] [Indexed: 02/07/2023] Open
Abstract
The bioenergetics of the vast majority of terrestrial mammals evolved to consuming glucose (Glc) for energy production under regular atmosphere (about 21% oxygen). However, some vertebrate species, such as aquatic turtles, seals, naked mole rat, and blind mole rat, Spalax, have adjusted their homeostasis to continuous function under severe hypoxic environment. The exploration of hypoxia-tolerant species metabolic strategies provides a better understanding of the adaptation to hypoxia. In this study, we compared Glc homeostasis in primary Spalax and rat skin cells under normoxic and hypoxic conditions. We used the targeted-metabolomics approach, utilizing liquid chromatography and mass spectrometry (LC-MS) to track the fate of heavy Glc carbons (13C6 Glc), as well as other methodologies to assist the interpretation of the metabolic landscape, such as bioenergetics profiling, Western blotting, and gene expression analysis. The metabolic profile was recorded under steady-state (after 24 h) of the experiment. Glc-originated carbons were unequally distributed between the cytosolic and mitochondrial domains in Spalax cells compared to the rat. The cytosolic domain is dominant apparently due to the hypoxia-inducible factor-1 alpha (HIF-1α) mastering, since its level is higher under normoxia and hypoxia in Spalax cells. Consumed Glc in Spalax cells is utilized for the pentose phosphate pathway maintaining the NADPH pool, and is finally harbored as glutathione (GSH) and UDP-GlcNAc. The cytosolic domain in Spalax cells works in the semi-uncoupled mode that limits the consumed Glc-derived carbons flux to the tricarboxylic acid (TCA) cycle and reduces pyruvate delivery; however, it maintains the NAD+ pool via lactate dehydrogenase upregulation. Both normoxic and hypoxic mitochondrial homeostasis of Glc-originated carbons in Spalax are characterized by their massive cataplerotic flux along with the axis αKG→Glu→Pro→hydroxyproline (HPro). The product of collagen degradation, HPro, as well as free Pro are apparently involved in the bioenergetics of Spalax under both normoxia and hypoxia. The upregulation of 2-hydroxyglutarate production detected in Spalax cells may be involved in modulating the levels of HIF-1α. Collectively, these data suggest that Spalax cells utilize similar metabolic frame for both normoxia and hypoxia, where glucose metabolism is switched from oxidative pathways (conversion of pyruvate to Acetyl-CoA and further TCA cycle processes) to (i) pentose phosphate pathway, (ii) lactate production, and (iii) cataplerotic pathways leading to hexosamine, GSH, and HPro production.
Collapse
Affiliation(s)
- Dmitry Miskevich
- Department of Evolutionary and Environmental Biology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel;
- Correspondence: (D.M.); (I.S.)
| | - Anastasia Chaban
- Department of Evolutionary and Environmental Biology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel;
| | - Maria Dronina
- Institute of Evolution, University of Haifa, Haifa 3498838, Israel;
| | - Ifat Abramovich
- Technion Faculty of Medicine, Haifa 3525433, Israel; (I.A.); (E.G.)
| | - Eyal Gottlieb
- Technion Faculty of Medicine, Haifa 3525433, Israel; (I.A.); (E.G.)
| | - Imad Shams
- Department of Evolutionary and Environmental Biology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel;
- Institute of Evolution, University of Haifa, Haifa 3498838, Israel;
- Correspondence: (D.M.); (I.S.)
| |
Collapse
|
50
|
Jia Q, Xie B, Zhao Z, Huang L, Wei G, Ni T. Lung cancer cells expressing a shortened CDK16 3'UTR escape senescence through impaired miR-485-5p targeting. Mol Oncol 2021; 16:1347-1364. [PMID: 34687270 PMCID: PMC8936527 DOI: 10.1002/1878-0261.13125] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 09/27/2021] [Accepted: 10/21/2021] [Indexed: 12/13/2022] Open
Abstract
Inducing senescence in cancer cells is an emerging strategy for cancer therapy. The dysregulation and mutation of genes encoding cyclin‐dependent kinases (CDKs) have been implicated in various human cancers. However, whether CDK can induce cancer cell senescence remains poorly understood. We observed that CDK16 expression was high in multiple cancer types, including lung cancer, whereas various replicative senescence models displayed low CDK16 expression. CDK16 knockdown caused senescence‐associated phenotypes in lung cancer cell lines. Interestingly, the CDK16 3′ UTR was shortened in cancer and lengthened in senescence models, which was regulated by alternative polyadenylation (APA). The longer 3′UTR [using the distal polyA (pA) site] generated less protein than the shorter one (using the proximal pA site). Since microRNAs (miRNAs) usually bind to the 3′UTR of target genes to suppress their expression, we investigated whether miRNAs targeting the region between the shortened and longer 3′UTR are responsible for the reduced expression. We found that miR‐485‐5p targeted the 3′UTR between the distal and proximal pA site and caused senescence‐associated phenotypes by reducing protein production from the longer CDK16 transcript. Of note, CDK16 knockdown led to a reduced expression of MYC proto‐oncogene, bHLH transcription factor (MYC) and CD274 molecule (PD‐L1), which in turn enhanced the tumor‐suppressive effects of senescent cancer cells. The present study discovered that CDK16, whose expression is under the regulation of APA and miR‐485‐5p, is a potential target for prosenescence therapy for lung cancer.
Collapse
Affiliation(s)
- Qi Jia
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, School of Life Sciences and Huashan Hospital, Fudan University, Shanghai, China
| | - Baiyun Xie
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, School of Life Sciences and Huashan Hospital, Fudan University, Shanghai, China
| | - Zhaozhao Zhao
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, School of Life Sciences and Huashan Hospital, Fudan University, Shanghai, China
| | - Leihuan Huang
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, School of Life Sciences and Huashan Hospital, Fudan University, Shanghai, China
| | - Gang Wei
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, School of Life Sciences and Huashan Hospital, Fudan University, Shanghai, China
| | - Ting Ni
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, School of Life Sciences and Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|