1
|
Kilili H, Padilla-Morales B, Castillo-Morales A, Monzón-Sandoval J, Díaz-Barba K, Cornejo-Paramo P, Vincze O, Giraudeau M, Bush SJ, Li Z, Chen L, Mourkas E, Ancona S, Gonzalez-Voyer A, Cortez D, Gutierrez H, Székely T, Acuña-Alonzo AP, Urrutia AO. Maximum lifespan and brain size in mammals are associated with gene family size expansion related to immune system functions. Sci Rep 2025; 15:15087. [PMID: 40301502 PMCID: PMC12041557 DOI: 10.1038/s41598-025-98786-3] [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: 09/17/2024] [Accepted: 04/15/2025] [Indexed: 05/01/2025] Open
Abstract
Mammals exhibit an unusual variation in their maximum lifespan potential, measured as the longest recorded longevity of any individual in a species. Evidence suggests that lifespan increases follow expansion in brain size relative to body mass. Here, we found significant gene family size expansions associated with maximum lifespan potential and relative brain size but not in gestation time, age of sexual maturity, and body mass in 46 mammalian species. Extended lifespan is associated with expanding gene families enriched in immune system functions. Our results suggest an association between gene duplication in immune-related gene families and the evolution of longer lifespans in mammals. These findings explore the genomic features linked with the evolution of lifespan in mammals and its association with life story and morphological traits.
Collapse
Affiliation(s)
- Huseyin Kilili
- Milner Centre for Evolution, Department of Life Sciences, University of Bath, Bath, BA2 7AY, UK
| | - Benjamin Padilla-Morales
- Milner Centre for Evolution, Department of Life Sciences, University of Bath, Bath, BA2 7AY, UK.
| | | | | | - Karina Díaz-Barba
- Milner Centre for Evolution, Department of Life Sciences, University of Bath, Bath, BA2 7AY, UK
- Instituto de Ecología, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico
- Licenciatura en Ciencias Genómicas, Universidad Nacional Autónoma de México, CP62210, Cuernavaca, Mexico
| | - Paola Cornejo-Paramo
- Milner Centre for Evolution, Department of Life Sciences, University of Bath, Bath, BA2 7AY, UK
- Licenciatura en Ciencias Genómicas, Universidad Nacional Autónoma de México, CP62210, Cuernavaca, Mexico
| | - Orsolya Vincze
- Littoral, Environnement et Sociétés (LIENSs), UMR 7266 CNRS-La Rochelle Université, 2 Rue Olympe de Gouges, FR-17000, La Rochelle, France
- Institute of Aquatic Ecology, Centre for Ecological Research, 4026, Debrecen, Hungary
- Evolutionary Ecology Group, Hungarian Department of Biology and Ecology, Babeş-Bolyai University, 400006, Cluj-Napoca, Romania
| | - Mathieu Giraudeau
- Littoral, Environnement et Sociétés (LIENSs), UMR 7266 CNRS-La Rochelle Université, 2 Rue Olympe de Gouges, FR-17000, La Rochelle, France
| | - Stephen J Bush
- School of Automation Science and Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Zhidan Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Laboratory Medicine, State Key Laboratory of Biotherapy, West China Second Hospital, Sichuan University, 610041, Chengdu, China
| | - Lu Chen
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Laboratory Medicine, State Key Laboratory of Biotherapy, West China Second Hospital, Sichuan University, 610041, Chengdu, China
| | - Evangelos Mourkas
- Zoonosis Science Centre, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Sergio Ancona
- Instituto de Ecología, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico
| | | | - Diego Cortez
- Centro de Ciencias Genómicas, Universidad Nacional Autónoma de México, CP62210, Cuernavaca, México
| | - Humberto Gutierrez
- Instituto Nacional de Medicina Genomica, 14610, Ciudad de Mexico, Mexico
| | - Tamás Székely
- Milner Centre for Evolution, Department of Life Sciences, University of Bath, Bath, BA2 7AY, UK
- Department of Evolutionary Zoology and Human Biology, University of Debrecen, Debrecen, Hungary
| | - Alín P Acuña-Alonzo
- Instituto de Ecología, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico.
| | - Araxi O Urrutia
- Milner Centre for Evolution, Department of Life Sciences, University of Bath, Bath, BA2 7AY, UK.
- Instituto de Ecología, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico.
| |
Collapse
|
2
|
Dos Santos Oliveira M, de C Griebeler M, Henz B, Dos Santos FF, Guardia GDA, Conceição HB, Galante PAF, Minussi DC, Oliveira MM, Lenz G. Population dynamics is a cancer driver. Carcinogenesis 2024; 45:893-902. [PMID: 38842162 DOI: 10.1093/carcin/bgae038] [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: 03/01/2024] [Revised: 05/16/2024] [Accepted: 06/04/2024] [Indexed: 06/07/2024] Open
Abstract
Most tissues are continuously renovated through the division of stem cells and the death of old or damaged cells, which is known as the cell turnover rate (CTOR). Despite being in a steady state, tissues have different population dynamics thus producing diverse clonality levels. Here, we propose and test that cell population dynamics can be a cancer driver. We employed the evolutionary software esiCancer to show that CTOR, within a range comparable to what is observed in human tissues, can amplify the risk of a mutation due to ancestral selection (ANSEL). In a high CTOR tissue, a mutated ancestral cell is likely to be selected and persist over generations, which leads to a scenario of elevated ANSEL profile, characterized by few niches of large clones, which does not occur in low CTOR. We found that CTOR is significantly associated with the risk of developing cancer, even when correcting for mutation load, indicating that population dynamics per se is a cancer driver. This concept is central to understanding cancer risk and for the design of new therapeutic interventions that minimizes the contribution of ANSEL in cancer growth.
Collapse
Affiliation(s)
- Mariana Dos Santos Oliveira
- Departamento de Biofísica, Universidade Federal do Rio Grande do Sul (UFRGS), Av. Bento Gonçalves, 9500, 91501-970, Porto Alegre, RS, Brazil
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), Av. Bento Gonçalves, 9500, 91501-970, Porto Alegre, RS, Brazil
| | - Marcelo de C Griebeler
- Departamento de Economia e Relações Internacionais, Universidade Federal do Rio Grande do Sul (UFRGS), Av. João Pessoa, 52, 90040-000, Porto Alegre, RS, Brazil
| | - Bernardo Henz
- Instituto de Informática, Universidade Federal do Rio Grande do Sul (UFRGS), Av. Bento Gonçalves, 9500, 91501-970, Porto Alegre, RS, Brazil
- Instituto Federal Farroupilha, Campus Alegrete, Rodovia RS-377, s/n, 97555-000 Alegrete, RS, Brazil
| | - Filipe Ferreira Dos Santos
- Centro de Oncologia Molecular, Hospital Sirio-Libanes, Prof Daher Cutait, 69, 013080-60, São Paulo, SP, Brazil
- Departamento de Bioquimica, Instituto de Quimica, Universidade de São Paulo, Av. Prof. Lineu Prestes, 748, 05508-090, São Paulo, SP, Brazil
| | - Gabriela D A Guardia
- Centro de Oncologia Molecular, Hospital Sirio-Libanes, Prof Daher Cutait, 69, 013080-60, São Paulo, SP, Brazil
| | - Helena B Conceição
- Centro de Oncologia Molecular, Hospital Sirio-Libanes, Prof Daher Cutait, 69, 013080-60, São Paulo, SP, Brazil
- Interunidades em Bioinformática, Universidade de São Paulo, R. do Matão, 1010, 05508-090, São Paulo, SP, Brazil
| | - Pedro A F Galante
- Centro de Oncologia Molecular, Hospital Sirio-Libanes, Prof Daher Cutait, 69, 013080-60, São Paulo, SP, Brazil
- Departamento de Bioquimica, Instituto de Quimica, Universidade de São Paulo, Av. Prof. Lineu Prestes, 748, 05508-090, São Paulo, SP, Brazil
| | - Darlan C Minussi
- Departamento de Biofísica, Universidade Federal do Rio Grande do Sul (UFRGS), Av. Bento Gonçalves, 9500, 91501-970, Porto Alegre, RS, Brazil
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), Av. Bento Gonçalves, 9500, 91501-970, Porto Alegre, RS, Brazil
| | - Manuel M Oliveira
- Instituto de Informática, Universidade Federal do Rio Grande do Sul (UFRGS), Av. Bento Gonçalves, 9500, 91501-970, Porto Alegre, RS, Brazil
| | - Guido Lenz
- Departamento de Biofísica, Universidade Federal do Rio Grande do Sul (UFRGS), Av. Bento Gonçalves, 9500, 91501-970, Porto Alegre, RS, Brazil
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), Av. Bento Gonçalves, 9500, 91501-970, Porto Alegre, RS, Brazil
| |
Collapse
|
3
|
Gao AW, El Alam G, Zhu Y, Li W, Sulc J, Li X, Katsyuba E, Li TY, Overmyer KA, Lalou A, Mouchiroud L, Sleiman MB, Cornaglia M, Morel JD, Houtkooper RH, Coon JJ, Auwerx J. High-content phenotypic analysis of a C. elegans recombinant inbred population identifies genetic and molecular regulators of lifespan. Cell Rep 2024; 43:114836. [PMID: 39368088 PMCID: PMC11996002 DOI: 10.1016/j.celrep.2024.114836] [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: 02/08/2024] [Revised: 07/10/2024] [Accepted: 09/20/2024] [Indexed: 10/07/2024] Open
Abstract
Lifespan is influenced by complex interactions between genetic and environmental factors. Studying those factors in model organisms of a single genetic background limits their translational value for humans. Here, we mapped lifespan determinants in 85 C. elegans recombinant inbred advanced intercross lines (RIAILs). We assessed molecular profiles-transcriptome, proteome, and lipidome-and life-history traits, including lifespan, development, growth dynamics, and reproduction. RIAILs exhibited large variations in lifespan, which correlated positively with developmental time. We validated three longevity modulators, including rict-1, gfm-1, and mltn-1, among the top candidates obtained from multiomics data integration and quantitative trait locus (QTL) mapping. We translated their relevance to humans using UK Biobank data and showed that variants in GFM1 are associated with an elevated risk of age-related heart failure. We organized our dataset as a resource that allows interactive explorations for new longevity targets.
Collapse
Affiliation(s)
- Arwen W Gao
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland; Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands.
| | - Gaby El Alam
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Yunyun Zhu
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53506, USA
| | - Weisha Li
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands
| | - Jonathan Sulc
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Xiaoxu Li
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Elena Katsyuba
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland; Nagi Bioscience SA, EPFL Innovation Park, 1025 Saint-Sulpice, Switzerland
| | - Terytty Y Li
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Katherine A Overmyer
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53506, USA; National Center for Quantitative Biology of Complex Systems, Madison, WI 53706, USA; Morgridge Institute for Research, Madison, WI 53515, USA
| | - Amelia Lalou
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Laurent Mouchiroud
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland; Nagi Bioscience SA, EPFL Innovation Park, 1025 Saint-Sulpice, Switzerland
| | - Maroun Bou Sleiman
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Matteo Cornaglia
- Nagi Bioscience SA, EPFL Innovation Park, 1025 Saint-Sulpice, Switzerland
| | - Jean-David Morel
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Riekelt H Houtkooper
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands
| | - Joshua J Coon
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53506, USA; National Center for Quantitative Biology of Complex Systems, Madison, WI 53706, USA; Morgridge Institute for Research, Madison, WI 53515, USA; Department of Chemistry, University of Wisconsin, Madison, WI 53506, USA
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland.
| |
Collapse
|
4
|
Jonischkies K, del Angel M, Demiray YE, Loaiza Zambrano A, Stork O. The NDR family of kinases: essential regulators of aging. Front Mol Neurosci 2024; 17:1371086. [PMID: 38803357 PMCID: PMC11129689 DOI: 10.3389/fnmol.2024.1371086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 04/22/2024] [Indexed: 05/29/2024] Open
Abstract
Aging is defined as a progressive decline of cognitive and physiological functions over lifetime. Since the definition of the nine hallmarks of aging in 2013 by López-Otin, numerous studies have attempted to identify the main regulators and contributors in the aging process. One interesting group of proteins whose participation has been implicated in several aging hallmarks are the nuclear DBF2-related (NDR) family of serine-threonine AGC kinases. They are one of the core components of the Hippo signaling pathway and include NDR1, NDR2, LATS1 and LATS2 in mammals, along with its highly conserved metazoan orthologs; Trc in Drosophila melanogaster, SAX-1 in Caenorhabditis elegans, CBK1, DBF20 in Saccharomyces cerevisiae and orb6 in Saccharomyces pombe. These kinases have been independently linked to the regulation of widely diverse cellular processes disrupted during aging such as the cell cycle progression, transcription, intercellular communication, nutrient homeostasis, autophagy, apoptosis, and stem cell differentiation. However, a comprehensive overview of the state-of-the-art knowledge regarding the post-translational modifications of and by NDR kinases in aging has not been conducted. In this review, we summarize the current understanding of the NDR family of kinases, focusing on their relevance to various aging hallmarks, and emphasize the growing body of evidence that suggests NDR kinases are essential regulators of aging across species.
Collapse
Affiliation(s)
- Kevin Jonischkies
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Miguel del Angel
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Yunus Emre Demiray
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Allison Loaiza Zambrano
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Oliver Stork
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Center for Behavioral Brain Science, Magdeburg, Germany
- Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Jena-Magdeburg-Halle, Germany
- German Center for Mental Health (DZPG), Jena-Magdeburg-Halle, Germany
| |
Collapse
|
5
|
Gao AW, Alam GE, Zhu Y, Li W, Katsyuba E, Sulc J, Li TY, Li X, Overmyer KA, Lalou A, Mouchiroud L, Sleiman MB, Cornaglia M, Morel JD, Houtkooper RH, Coon JJ, Auwerx J. High-content phenotypic analysis of a C. elegans recombinant inbred population identifies genetic and molecular regulators of lifespan. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.15.575638. [PMID: 38293129 PMCID: PMC10827074 DOI: 10.1101/2024.01.15.575638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Lifespan is influenced by complex interactions between genetic and environmental factors. Studying those factors in model organisms of a single genetic background limits their translational value for humans. Here, we mapped lifespan determinants in 85 genetically diverse C. elegans recombinant intercross advanced inbred lines (RIAILs). We assessed molecular profiles - transcriptome, proteome, and lipidome - and life-history traits, including lifespan, development, growth dynamics, and reproduction. RIAILs exhibited large variations in lifespan, which positively correlated with developmental time. Among the top candidates obtained from multi-omics data integration and QTL mapping, we validated known and novel longevity modulators, including rict-1, gfm-1 and mltn-1. We translated their relevance to humans using UK Biobank data and showed that variants in RICTOR and GFM1 are associated with an elevated risk of age-related heart disease, dementia, diabetes, kidney, and liver diseases. We organized our dataset as a resource (https://lisp-lms.shinyapps.io/RIAILs/) that allows interactive explorations for new longevity targets.
Collapse
Affiliation(s)
- Arwen W. Gao
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Gaby El Alam
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Yunyun Zhu
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53506, USA
| | - Weisha Li
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Elena Katsyuba
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
- Nagi Bioscience SA, EPFL Innovation Park, CH-1025 Saint-Sulpice, Switzerland
| | - Jonathan Sulc
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Terytty Y. Li
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
- Present address: State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Metabolic Remodeling and Health, Laboratory of Longevity and Metabolic Adaptations, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Xiaoxu Li
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Katherine A. Overmyer
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53506, USA
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53706, USA
- Morgridge Institute for Research, Madison, WI 53515, USA
| | - Amelia Lalou
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Laurent Mouchiroud
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
- Nagi Bioscience SA, EPFL Innovation Park, CH-1025 Saint-Sulpice, Switzerland
| | - Maroun Bou Sleiman
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Matteo Cornaglia
- Nagi Bioscience SA, EPFL Innovation Park, CH-1025 Saint-Sulpice, Switzerland
| | - Jean-David Morel
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Riekelt H. Houtkooper
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Joshua J. Coon
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53506, USA
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53706, USA
- Morgridge Institute for Research, Madison, WI 53515, USA
- Department of Chemistry, University of Wisconsin, Madison, WI 53506, USA
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| |
Collapse
|
6
|
Yang M, Harrison BR, Promislow DEL. Cellular age explains variation in age-related cell-to-cell transcriptome variability. Genome Res 2023; 33:1906-1916. [PMID: 37973195 PMCID: PMC10760448 DOI: 10.1101/gr.278144.123] [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: 05/31/2023] [Accepted: 10/12/2023] [Indexed: 11/19/2023]
Abstract
Organs and tissues age at different rates within a single individual. Such asynchrony in aging has been widely observed at multiple levels, from functional hallmarks, such as anatomical structures and physiological processes, to molecular endophenotypes, such as the transcriptome and metabolome. However, we lack a conceptual framework to understand why some components age faster than others. Just as demographic models explain why aging evolves, here we test the hypothesis that demographic differences among cell types, determined by cell-specific differences in turnover rate, can explain why the transcriptome shows signs of aging in some cell types but not others. Through analysis of mouse single-cell transcriptome data across diverse tissues and ages, we find that cellular age explains a large proportion of the variation in the age-related increase in transcriptome variance. We further show that long-lived cells are characterized by relatively high expression of genes associated with proteostasis and that the transcriptome of long-lived cells shows greater evolutionary constraint than short-lived cells. In contrast, in short-lived cell types, the transcriptome is enriched for genes associated with DNA repair. Based on these observations, we develop a novel heuristic model that explains how and why aging rates differ among cell types.
Collapse
Affiliation(s)
- Ming Yang
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington 98195, USA
| | - Benjamin R Harrison
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington 98195, USA
| | - Daniel E L Promislow
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington 98195, USA;
- Department of Biology, University of Washington, Seattle, Washington 98195, USA
| |
Collapse
|
7
|
Yi S, Wong RKW, Gaynanova I. Hierarchical nuclear norm penalization for multi-view data integration. Biometrics 2023; 79:2933-2946. [PMID: 37345491 DOI: 10.1111/biom.13893] [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: 06/24/2022] [Accepted: 05/18/2023] [Indexed: 06/23/2023]
Abstract
The prevalence of data collected on the same set of samples from multiple sources (i.e., multi-view data) has prompted significant development of data integration methods based on low-rank matrix factorizations. These methods decompose signal matrices from each view into the sum of shared and individual structures, which are further used for dimension reduction, exploratory analyses, and quantifying associations across views. However, existing methods have limitations in modeling partially-shared structures due to either too restrictive models, or restrictive identifiability conditions. To address these challenges, we propose a new formulation for signal structures that include partially-shared signals based on grouping the views into so-called hierarchical levels with identifiable guarantees under suitable conditions. The proposed hierarchy leads us to introduce a new penalty, hierarchical nuclear norm (HNN), for signal estimation. In contrast to existing methods, HNN penalization avoids scores and loadings factorization of the signals and leads to a convex optimization problem, which we solve using a dual forward-backward algorithm. We propose a simple refitting procedure to adjust the penalization bias and develop an adapted version of bi-cross-validation for selecting tuning parameters. Extensive simulation studies and analysis of the genotype-tissue expression data demonstrate the advantages of our method over existing alternatives.
Collapse
Affiliation(s)
- Sangyoon Yi
- Department of Statistics, Oklahoma State University, Stillwater, Oklahoma, USA
| | | | - Irina Gaynanova
- Department of Statistics, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
8
|
Alberry B, Silveira PP. Brain insulin signaling as a potential mediator of early life adversity effects on physical and mental health. Neurosci Biobehav Rev 2023; 153:105350. [PMID: 37544390 DOI: 10.1016/j.neubiorev.2023.105350] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 07/31/2023] [Accepted: 08/03/2023] [Indexed: 08/08/2023]
Abstract
In numerous brain structures, insulin signaling modulates the homeostatic processes, sensitivity to reward pathways, executive function, memory, and cognition. Through human studies and animal models, mounting evidence implicates central insulin signaling in the metabolic, physiological, and psychological consequences of early life adversity. In this review, we describe the consequences of early life adversity in the brain where insulin signaling is a key factor and how insulin may moderate the effects of adversity on psychiatric and cardio-metabolic health outcomes. Further understanding of how early life adversity and insulin signaling impact specific brain regions and mental and physical health outcomes will assist in prevention, diagnosis, and potential intervention following early life adversity.
Collapse
Affiliation(s)
- Bonnie Alberry
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Patricia Pelufo Silveira
- Department of Psychiatry, McGill University, Montreal, QC, Canada; Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada.
| |
Collapse
|
9
|
Lamichhane PP, Samir P. Cellular Stress: Modulator of Regulated Cell Death. BIOLOGY 2023; 12:1172. [PMID: 37759572 PMCID: PMC10525759 DOI: 10.3390/biology12091172] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/22/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023]
Abstract
Cellular stress response activates a complex program of an adaptive response called integrated stress response (ISR) that can allow a cell to survive in the presence of stressors. ISR reprograms gene expression to increase the transcription and translation of stress response genes while repressing the translation of most proteins to reduce the metabolic burden. In some cases, ISR activation can lead to the assembly of a cytoplasmic membraneless compartment called stress granules (SGs). ISR and SGs can inhibit apoptosis, pyroptosis, and necroptosis, suggesting that they guard against uncontrolled regulated cell death (RCD) to promote organismal homeostasis. However, ISR and SGs also allow cancer cells to survive in stressful environments, including hypoxia and during chemotherapy. Therefore, there is a great need to understand the molecular mechanism of the crosstalk between ISR and RCD. This is an active area of research and is expected to be relevant to a range of human diseases. In this review, we provided an overview of the interplay between different cellular stress responses and RCD pathways and their modulation in health and disease.
Collapse
Affiliation(s)
| | - Parimal Samir
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
10
|
Mposhi A, Turner JD. How can early life adversity still exert an effect decades later? A question of timing, tissues and mechanisms. Front Immunol 2023; 14:1215544. [PMID: 37457711 PMCID: PMC10348484 DOI: 10.3389/fimmu.2023.1215544] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 06/14/2023] [Indexed: 07/18/2023] Open
Abstract
Exposure to any number of stressors during the first 1000 days from conception to age 2 years is important in shaping an individual's life trajectory of health and disease. Despite the expanding range of stressors as well as later-life phenotypes and outcomes, the underlying molecular mechanisms remain unclear. Our previous data strongly suggests that early-life exposure to a stressor reduces the capacity of the immune system to generate subsequent generations of naïve cells, while others have shown that, early life stress impairs the capacity of neuronal stem cells to proliferate as they age. This leads us to the "stem cell hypothesis" whereby exposure to adversity during a sensitive period acts through a common mechanism in all the cell types by programming the tissue resident progenitor cells. Furthermore, we review the mechanistic differences observed in fully differentiated cells and suggest that early life adversity (ELA) may alter mitochondria in stem cells. This may consequently alter the destiny of these cells, producing the lifelong "supply" of functionally altered fully differentiated cells.
Collapse
|
11
|
Rigby Dames BA, Kilili H, Charvet CJ, Díaz-Barba K, Proulx MJ, de Sousa AA, Urrutia AO. Evolutionary and genomic perspectives of brain aging and neurodegenerative diseases. PROGRESS IN BRAIN RESEARCH 2023; 275:165-215. [PMID: 36841568 PMCID: PMC11191546 DOI: 10.1016/bs.pbr.2022.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
This chapter utilizes genomic concepts and evolutionary perspectives to further understand the possible links between typical brain aging and neurodegenerative diseases, focusing on the two most prevalent of these: Alzheimer's disease and Parkinson's disease. Aging is the major risk factor for these neurodegenerative diseases. Researching the evolutionary and molecular underpinnings of aging helps to reveal elements of the typical aging process that leave individuals more vulnerable to neurodegenerative pathologies. Very little is known about the prevalence and susceptibility of neurodegenerative diseases in nonhuman species, as only a few individuals have been observed with these neuropathologies. However, several studies have investigated the evolution of lifespan, which is closely connected with brain size in mammals, and insights can be drawn from these to enrich our understanding of neurodegeneration. This chapter explores the relationship between the typical aging process and the events in neurodegeneration. First, we examined how age-related processes can increase susceptibility to neurodegenerative diseases. Second, we assessed to what extent neurodegeneration is an accelerated form of aging. We found that while at the phenotypic level both neurodegenerative diseases and the typical aging process share some characteristics, at the molecular level they show some distinctions in their profiles, such as variation in genes and gene expression. Furthermore, neurodegeneration of the brain is associated with an earlier onset of cellular, molecular, and structural age-related changes. In conclusion, a more integrative view of the aging process, both from a molecular and an evolutionary perspective, may increase our understanding of neurodegenerative diseases.
Collapse
Affiliation(s)
- Brier A Rigby Dames
- Department of Computer Science, University of Bath, Bath, United Kingdom; Department of Psychology, University of Bath, Bath, United Kingdom.
| | - Huseyin Kilili
- Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Christine J Charvet
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Karina Díaz-Barba
- Licenciatura en Ciencias Genómicas, UNAM, CP62210, Cuernavaca, México; Instituto de Ecología, UNAM, Ciudad Universitaria, CP04510, Ciudad de México, México
| | - Michael J Proulx
- Department of Psychology, University of Bath, Bath, United Kingdom
| | | | - Araxi O Urrutia
- Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom; Licenciatura en Ciencias Genómicas, UNAM, CP62210, Cuernavaca, México; Instituto de Ecología, UNAM, Ciudad Universitaria, CP04510, Ciudad de México, México.
| |
Collapse
|
12
|
Mota-Martorell N, Jové M, Berdún R, Òbis È, Barja G, Pamplona R. Methionine Metabolism Is Down-Regulated in Heart of Long-Lived Mammals. BIOLOGY 2022; 11:biology11121821. [PMID: 36552330 PMCID: PMC9775425 DOI: 10.3390/biology11121821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/11/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
Methionine constitutes a central hub of intracellular metabolic adaptations leading to an extended longevity (maximum lifespan). The present study follows a comparative approach analyzing methionine and related metabolite and amino acid profiles using an LC-MS/MS platform in the hearts of seven mammalian species with a longevity ranging from 3.8 to 57 years. Our findings demonstrate the existence of species-specific heart phenotypes associated with high longevity characterized by: (i) low concentration of methionine and its related sulphur-containing metabolites; (ii) low amino acid pool; and (iii) low choline concentration. Our results support the existence of heart metabotypes characterized by a down-regulation in long-lived species, supporting the idea that in longevity, less is more.
Collapse
Affiliation(s)
- Natalia Mota-Martorell
- Department of Experimental Medicine, University of Lleida-Biomedical Research Institute of Lleida (UdL-IRBLleida), 25008 Lleida, Spain
| | - Mariona Jové
- Department of Experimental Medicine, University of Lleida-Biomedical Research Institute of Lleida (UdL-IRBLleida), 25008 Lleida, Spain
| | - Rebeca Berdún
- Department of Experimental Medicine, University of Lleida-Biomedical Research Institute of Lleida (UdL-IRBLleida), 25008 Lleida, Spain
| | - Èlia Òbis
- Department of Experimental Medicine, University of Lleida-Biomedical Research Institute of Lleida (UdL-IRBLleida), 25008 Lleida, Spain
| | - Gustavo Barja
- Department of Genetics, Physiology and Microbiology, Complutense University, 28040 Madrid, Spain
| | - Reinald Pamplona
- Department of Experimental Medicine, University of Lleida-Biomedical Research Institute of Lleida (UdL-IRBLleida), 25008 Lleida, Spain
- Correspondence:
| |
Collapse
|
13
|
Nitta RT, Luo EJ, Lim M, Li G. Can tumor treating fields induce DNA damage and reduce cell motility in medulloblastoma cell lines? J Neurosurg Pediatr 2022; 30:555-566. [PMID: 36208441 DOI: 10.3171/2022.8.peds22300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/25/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Medulloblastoma (MB) is the most common malignant pediatric brain tumor and accounts for approximately 20% of all pediatric CNS tumors. Current multimodal treatment is associated with a 70%-90% 5-year survival rate; however, the prognosis for patients with tumor dissemination and recurrent MB remains poor. The majority of survivors exhibit long-term neurocognitive complications; thus, more effective and less toxic treatments are critically needed. Tumor treating fields (TTFields) are low-intensity, alternating electric fields that disrupt cell division through physical interactions with key molecules during mitosis. Side effects from TTField therapy are minimal, making it an ideal candidate for MB treatment. METHODS To determine if TTFields can be an effective treatment for MB, the authors conducted an in vitro study treating multiple MB cell lines. Three MB molecular subgroups (SHH [sonic hedgehog], group 3, and group 4) were treated for 24, 48, and 72 hours at 100, 200, 300, and 400 kHz. Combinatorial studies were conducted with the small-molecule casein kinase 2 inhibitor CX-4945. RESULTS TTFields reduced MB cell growth with an optimal frequency of 300 kHz, and the most efficacious treatment time was 72 hours. Treatment with TTFields dysregulated actin polymerization and corresponded with a reduction in cell motility and invasion. TTFields also induced DNA damage (γH2AX, 53BP1) that correlated with an increase in apoptotic cells. The authors discovered that CX-4945 works synergistically with TTFields to reduce MB growth. In addition, combining CX-4945 and TTFields increased the cellular actin dysregulation, which correlated with a decrease in MB migration. CONCLUSIONS The findings of this study demonstrate that TTFields may be a novel and less toxic method to treat patients with MB.
Collapse
|
14
|
Caloric restriction reinforces the stem cell pool in the aged brain without affecting overall proliferation status. Gene X 2022; 851:147026. [DOI: 10.1016/j.gene.2022.147026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 09/21/2022] [Accepted: 10/27/2022] [Indexed: 11/08/2022] Open
|
15
|
Silencing of the Ca2+ Channel ORAI1 Improves the Multi-Systemic Phenotype of Tubular Aggregate Myopathy (TAM) and Stormorken Syndrome (STRMK) in Mice. Int J Mol Sci 2022; 23:ijms23136968. [PMID: 35805973 PMCID: PMC9266658 DOI: 10.3390/ijms23136968] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/17/2022] [Accepted: 06/20/2022] [Indexed: 12/04/2022] Open
Abstract
Tubular aggregate myopathy (TAM) and Stormorken syndrome (STRMK) form a clinical continuum associating progressive muscle weakness with additional multi-systemic anomalies of the bones, skin, spleen, and platelets. TAM/STRMK arises from excessive extracellular Ca2+ entry due to gain-of-function mutations in the Ca2+ sensor STIM1 or the Ca2+ channel ORAI1. Currently, no treatment is available. Here we assessed the therapeutic potential of ORAI1 downregulation to anticipate and reverse disease development in a faithful mouse model carrying the most common TAM/STRMK mutation and recapitulating the main signs of the human disorder. To this aim, we crossed Stim1R304W/+ mice with Orai1+/− mice expressing 50% of ORAI1. Systematic phenotyping of the offspring revealed that the Stim1R304W/+Orai1+/− mice were born with a normalized ratio and showed improved postnatal growth, bone architecture, and partly ameliorated muscle function and structure compared with their Stim1R304W/+ littermates. We also produced AAV particles containing Orai1-specific shRNAs, and intramuscular injections of Stim1R304W/+ mice improved the skeletal muscle contraction and relaxation properties, while muscle histology remained unchanged. Altogether, we provide the proof-of-concept that Orai1 silencing partially prevents the development of the multi-systemic TAM/STRMK phenotype in mice, and we also established an approach to target Orai1 expression in postnatal tissues.
Collapse
|
16
|
Kumar AV, Kang T, Thakurta TG, Ng C, Rogers AN, Larsen MR, Lapierre LR. Exportin 1 modulates life span by regulating nucleolar dynamics via the autophagy protein LGG-1/GABARAP. SCIENCE ADVANCES 2022; 8:eabj1604. [PMID: 35363528 PMCID: PMC10938577 DOI: 10.1126/sciadv.abj1604] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 02/08/2022] [Indexed: 06/14/2023]
Abstract
Altered nucleolar and ribosomal dynamics are key hallmarks of aging, but their regulation remains unclear. Building on the knowledge that the conserved nuclear export receptor Exportin 1 (XPO-1/XPO1) modulates proteostasis and life span, we systematically analyzed the impact of nuclear export on protein metabolism. Using transcriptomic and subcellular proteomic analyses in nematodes, we demonstrate that XPO-1 modulates the nucleocytoplasmic distribution of key proteins involved in nucleolar dynamics and ribosome function, including fibrillarin (FIB-1/FBL) and RPL-11 (RPL11). Silencing xpo-1 led to marked reduction in global translation, which was accompanied by decreased nucleolar size and lower fibrillarin levels. A targeted screen of known proteostatic mediators revealed that the autophagy protein LGG-1/GABARAP modulates nucleolar size by regulating RPL-11 levels, linking specific protein degradation to ribosome metabolism. Together, our study reveals that nucleolar size and life span are regulated by LGG-1/GABARAP via ribosome protein surveillance.
Collapse
Affiliation(s)
- Anita V. Kumar
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, 185 Meeting St., Providence, RI 02912, USA
| | - Taewook Kang
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Tara G. Thakurta
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, 185 Meeting St., Providence, RI 02912, USA
| | - Celeste Ng
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, 185 Meeting St., Providence, RI 02912, USA
| | - Aric N. Rogers
- MDI Biological Laboratory, 159 Old Bar Harbor Rd., Salisbury Cove, ME 04672, USA
| | - Martin R. Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Louis R. Lapierre
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, 185 Meeting St., Providence, RI 02912, USA
| |
Collapse
|
17
|
Bruic M, Grujic-Milanovic J, Miloradovic Z, Jovovic D, Zivkovic L, Mihailovic-Stanojevic N, Karanovic D, Spremo-Potparevic B. DNA, protein and lipid oxidative damage in tissues of spontaneously hypertensive versus normotensive rats. Int J Biochem Cell Biol 2021; 141:106088. [PMID: 34601089 DOI: 10.1016/j.biocel.2021.106088] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 09/06/2021] [Accepted: 09/25/2021] [Indexed: 10/20/2022]
Abstract
Oxidative damage to protein and lipid macromolecules in target organs in hypertension has been recognized as a major factor contributing to cardiovascular, cerebrovascular, and renal diseases. Data on protein and lipid oxidative damage in spontaneously hypertensive rats are numerous, but there is no information on DNA damage in tissues measured by comet assay. The aim of this study was to determine the baseline damage to DNA, protein, and lipid macromolecules in different organs of spontaneously hypertensive rats. Markers of lipid peroxidation, protein oxidation, and DNA damage were measured in blood, heart, kidney, and liver of 24-week-old spontaneously hypertensive rats. Plasma prooxidant and antioxidant status were determined as well. Age-matched normotensive Wistar rats were used as control. A rise in markers of lipid peroxidation and protein oxidation, malondialdehyde, and advanced oxidation protein products, was detected in all tissues of spontaneously hypertensive rats, with particularly high values in the liver. DNA damage, measured by the comet assay, was significantly higher in all the studied tissues of spontaneously hypertensive rats compared to normotensive control, with more severe damage in the cardiac and renal cells. Significant depletion of the plasma antioxidant barrier in spontaneously hypertensive rats was also observed. This study showed increased damage to all macromolecules in all studied samples of spontaneously hypertensive rats in comparison with control Wistar rats.
Collapse
Affiliation(s)
- Marija Bruic
- Department of Pathobiology, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia.
| | - Jelica Grujic-Milanovic
- Department of Cardiovascular Physiology, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Zoran Miloradovic
- Department of Cardiovascular Physiology, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Djurdjica Jovovic
- Department of Cardiovascular Physiology, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Lada Zivkovic
- Department of Pathobiology, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Nevena Mihailovic-Stanojevic
- Department of Cardiovascular Physiology, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Danijela Karanovic
- Department of Cardiovascular Physiology, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | | |
Collapse
|
18
|
Blatt R, Davidi S, Munster M, Shteingauz A, Cahal S, Zeidan A, Marciano T, Bomzon Z, Haber A, Giladi M, Weinberg U, Kinzel A, Palti Y. In Vivo Safety of Tumor Treating Fields (TTFields) Applied to the Torso. Front Oncol 2021; 11:670809. [PMID: 34249709 PMCID: PMC8264759 DOI: 10.3389/fonc.2021.670809] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 06/09/2021] [Indexed: 12/24/2022] Open
Abstract
Background Tumor Treating Fields (TTFields) therapy is a non-invasive, loco-regional, anti-mitotic treatment modality that targets rapidly dividing cancerous cells, utilizing low intensity, alternating electric fields at cancer-cell-type specific frequencies. TTFields therapy is approved for the treatment of newly diagnosed and recurrent glioblastoma (GBM) in the US, Europe, Israel, Japan, and China. The favorable safety profile of TTFields in patients with GBM is partially attributed to the low rate of mitotic events in normal, quiescent brain cells. However, specific safety evaluations are warranted at locations with known high rates of cellular proliferation, such as the torso, which is a primary site of several of the most aggressive malignant tumors. Methods The safety of delivering TTFields to the torso of healthy rats at 150 or 200 kHz, which were previously identified as optimal frequencies for treating multiple torso cancers, was investigated. Throughout 2 weeks of TTFields application, animals underwent daily clinical examinations, and at treatment cessation blood samples and internal organs were examined. Computer simulations were performed to verify that the targeted internal organs of the torso were receiving TTFields at therapeutic intensities (≥ 1 V/cm root mean square, RMS). Results No treatment-related mortality was observed. Furthermore, no significant differences were observed between the TTFields-treated and control animals for all examined safety parameters: activity level, food and water intake, stools, motor neurological status, respiration, weight, complete blood count, blood biochemistry, and pathological findings of internal organs. TTFields intensities of 1 to 2.5 V/cm RMS were confirmed for internal organs within the target region. Conclusions This research demonstrates the safety of therapeutic level TTFields at frequencies of 150 and 200 kHz when applied as monotherapy to the torso of healthy rats.
Collapse
|
19
|
Kao HJ, Wang YH, Keshari S, Yang JJ, Simbolon S, Chen CC, Huang CM. Propionic acid produced by Cutibacterium acnes fermentation ameliorates ultraviolet B-induced melanin synthesis. Sci Rep 2021; 11:11980. [PMID: 34099789 PMCID: PMC8184931 DOI: 10.1038/s41598-021-91386-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 05/18/2021] [Indexed: 12/13/2022] Open
Abstract
Ultraviolet irradiation induces melanin accumulation, which can be reduced by the use of chemical whitening products. However, the associated safety concerns of such products have prompted the search for natural and harmless alternatives. This study aimed to identify a natural acidic formulation to reduce skin pigmentation. The metabolite propionic acid (CH3CH2COOH, PA) was the most abundant fatty acid in the filtrate from Pluronic F68 (PF68) fermentation of Cutibacterium acnes (C. acnes) and reduced the DOPA-positive melanocytes by significantly inhibiting cellular tyrosinase activity via binding to the free fatty acid receptor 2 (FFAR2). Moreover, 4 mM PA treatment did not alter melanocyte proliferation, indicating that it is an effective solution for hyperpigmentation, causing no cellular damage. The reduced DOPA-positive melanocytes and tyrosinase activity were also observed in mice ear skin tissue injected with a mixture of C. acnes and PF68, supporting that the inhibition of melanogenesis is likely to be mediated through fermentation metabolites from C. acnes fermentation using PF68 as a carbon source. Additionally, PA did not affect the growth of its parent bacteria C. acnes, hence is a potent fermentation metabolite that does not disrupt the balance of the skin microbiome.
Collapse
Affiliation(s)
- Hsin-Jou Kao
- Department of Biomedical Sciences and Engineering, National Central University, Zhongda Rd, No. 300, Zhongda Rd., Zhongli District, Taoyuan City, 32001, Taiwan, ROC
| | - Yan-Han Wang
- Department of Dermatology, University of California, San Diego, CA, USA
| | - Sunita Keshari
- Department of Life Sciences, National Central University, Zhongli District, Taoyuan City, Taiwan, ROC
| | - John Jackson Yang
- Department of Life Sciences, National Central University, Zhongli District, Taoyuan City, Taiwan, ROC
| | - Shinta Simbolon
- Department of Biomedical Sciences and Engineering, National Central University, Zhongda Rd, No. 300, Zhongda Rd., Zhongli District, Taoyuan City, 32001, Taiwan, ROC
| | - Chun-Chuan Chen
- Department of Biomedical Sciences and Engineering, National Central University, Zhongda Rd, No. 300, Zhongda Rd., Zhongli District, Taoyuan City, 32001, Taiwan, ROC
| | - Chun-Ming Huang
- Department of Biomedical Sciences and Engineering, National Central University, Zhongda Rd, No. 300, Zhongda Rd., Zhongli District, Taoyuan City, 32001, Taiwan, ROC.
| |
Collapse
|
20
|
Rodriguez D, Taketa DA, Madhu R, Kassmer S, Loerke D, Valentine MT, Tomaso AWD. Vascular Aging in the Invertebrate Chordate, Botryllus schlosseri. Front Mol Biosci 2021; 8:626827. [PMID: 33898513 PMCID: PMC8060491 DOI: 10.3389/fmolb.2021.626827] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 03/03/2021] [Indexed: 12/13/2022] Open
Abstract
Vascular diseases affect over 1 billion people worldwide and are highly prevalent among the elderly, due to a progressive deterioration of the structure of vascular cells. Most of our understanding of these age-related cellular changes comes from in vitro studies on human cell lines. Further studies of the mechanisms underlying vascular aging in vivo are needed to provide insight into the pathobiology of age-associated vascular diseases, but are difficult to carry out on vertebrate model organisms. We are studying the effects of aging on the vasculature of the invertebrate chordate, Botryllus schlosseri. This extracorporeal vascular network of Botryllus is transparent and particularly amenable to imaging and manipulation. Here we use a combination of transcriptomics, immunostaining and live-imaging, as well as in vivo pharmacological treatments and regeneration assays to show that morphological, transcriptional, and functional age-associated changes within vascular cells are key hallmarks of aging in B. schlosseri, and occur independent of genotype. We show that age-associated changes in the cytoskeleton and the extracellular matrix reshape vascular cells into a flattened and elongated form and there are major changes in the structure of the basement membrane over time. The vessels narrow, reducing blood flow, and become less responsive to stimuli inducing vascular regression. The extracorporeal vasculature is highly regenerative following injury, and while age does not affect the regeneration potential, newly regenerated vascular cells maintain the same aged phenotype, suggesting that aging of the vasculature is a result of heritable epigenetic changes.
Collapse
Affiliation(s)
- Delany Rodriguez
- Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Daryl A. Taketa
- Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Roopa Madhu
- Department of Physics and Astronomy, University of Denver, Denver, CO, United States
| | - Susannah Kassmer
- Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Dinah Loerke
- Department of Physics and Astronomy, University of Denver, Denver, CO, United States
| | - Megan T. Valentine
- Department of Mechanical Engineering, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Anthony W. De Tomaso
- Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA, United States
| |
Collapse
|
21
|
Erbaba B, Arslan-Ergul A, Adams MM. Effects of caloric restriction on the antagonistic and integrative hallmarks of aging. Ageing Res Rev 2021; 66:101228. [PMID: 33246078 DOI: 10.1016/j.arr.2020.101228] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 11/09/2020] [Accepted: 11/19/2020] [Indexed: 12/19/2022]
Abstract
Aging is a significant risk factor for cognitive decline associated with neurodegenerative diseases, which makes understanding what promotes 'healthy brain aging' very important. Studies suggest that caloric restriction (CR) is a non-genetic intervention that reliably extends life- and healthspan. Here, we review the CR literature related to both the subject of aging and alterations in cell cycle machinery, especially surrounding the regulation of the E2F/DP1 complex, to elucidate the cellular protection mechanisms in the brain induced via dietary applications. The alterations extending lifespan via CR appear to exert their effects by promoting survival of individual cells, downregulating cell proliferation, and inducing stem cell quiescence, which results in keeping the stem cell reserve for extreme needs. This survival instinct of cells is believed to cause some molecular adaptations for their maintenance of the system. Avoiding energy waste of proliferation machinery promotes the long term survival of the individual cells and this is due to adaptations to the limited nutrient supply in the environment. Such a protective mechanism induced by diet could be promoted via the downregulation of crucial cell cycle-related transcription activators. This review article aims to bring attention to the importance of molecular adaptations induced by diet that promote healthy brain aging. It will provide insights into alternative targets for new treatments or neuroprotective approaches against neurodegenerative pathophysiologies.
Collapse
Affiliation(s)
- Begun Erbaba
- Interdisciplinary Graduate Program in Neuroscience, Aysel Sabuncu Brain Research Center, Bilkent University, Ankara, Turkey; National Nanotechnology Research Center (UNAM), Bilkent University, Ankara, Turkey
| | - Ayca Arslan-Ergul
- National Nanotechnology Research Center (UNAM), Bilkent University, Ankara, Turkey
| | - Michelle M Adams
- Interdisciplinary Graduate Program in Neuroscience, Aysel Sabuncu Brain Research Center, Bilkent University, Ankara, Turkey; National Nanotechnology Research Center (UNAM), Bilkent University, Ankara, Turkey; Department of Psychology, Bilkent University, Ankara, Turkey.
| |
Collapse
|
22
|
Wang X, Bootsma H, Terpstra J, Vissink A, van der Vegt B, Spijkervet FKL, Kroese FGM, Pringle S. Progenitor cell niche senescence reflects pathology of the parotid salivary gland in primary Sjögren's syndrome. Rheumatology (Oxford) 2021; 59:3003-3013. [PMID: 32159757 PMCID: PMC7516109 DOI: 10.1093/rheumatology/keaa012] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 12/20/2019] [Indexed: 12/13/2022] Open
Abstract
Objective Salivary gland (SG) progenitor cells (SGPCs) maintain SG homeostasis. We have previously shown that in primary Sjögren’s syndrome (pSS), SGPCs are likely to be senescent, and may underpin SG dysfunction. This study assessed the extent of senescence of cells in a SGPC niche in pSS patients’ SGs, and its correlation with functional and clinical parameters. Methods The expression of p16 and p21 as markers of senescence in both total SG epithelium and a SGPC niche (basal striated duct cells, BSD) was examined in SGs of pSS (n = 35), incomplete pSS (n = 13) (patients with some signs of pSS, but not fulfilling all classification criteria) and non-SS sicca control (n = 21) patients. This was correlated with functional and clinical parameters. Results pSS patient SGs contained significantly more p16+ cells both in the epithelium in general (P <0.01) and in the BSD layer (P <0.001), than non-SS SGs. Significant correlations were found in pSS patients between p16+ BSD cells and secretion of unstimulated whole saliva, stimulated whole saliva, stimulated parotid saliva, CD45+ infiltrate, ultrasound total score and ACR-EULAR classification score, but not with EULAR Sjögren’s syndrome disease activity index (ESSDAI) and EULAR Sjögren’s Syndrome Patient Reported Index (ESSPRI) scores. Correlations with total epithelium p16+ cells were weaker. Incomplete pSS patients also had increased numbers of p16+ epithelial and BSD cells. Based on protein and mRNA expression, p21+ appears not to play a significant role in the SG in pSS. Conclusion These findings suggest SGPC senescence may be an early feature of primary Sjögren’s syndrome and may contribute to defective SG function in pSS but not to systemic disease activity.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Department of Rheumatology and Clinical ImmunologyUniversity Medical Center Groningen, Groningen, The Netherlands
| | - Hendrika Bootsma
- Department of Rheumatology and Clinical ImmunologyUniversity Medical Center Groningen, Groningen, The Netherlands
| | - Janneke Terpstra
- Department of Rheumatology and Clinical ImmunologyUniversity Medical Center Groningen, Groningen, The Netherlands
| | - Arjan Vissink
- Department of Oral and Maxillofacial Surgery, University Medical Center Groningen, Groningen, The Netherlands
| | - Bert van der Vegt
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Fred K L Spijkervet
- Department of Oral and Maxillofacial Surgery, University Medical Center Groningen, Groningen, The Netherlands
| | - Frans G M Kroese
- Department of Rheumatology and Clinical ImmunologyUniversity Medical Center Groningen, Groningen, The Netherlands
| | - Sarah Pringle
- Department of Rheumatology and Clinical ImmunologyUniversity Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
23
|
Li C, Nong Q, Guan B, He H, Zhang Z. Specific Differentially Methylated and Expressed Genes in People with Longevity Family History. IRANIAN JOURNAL OF PUBLIC HEALTH 2021; 50:152-160. [PMID: 34178774 PMCID: PMC8213620 DOI: 10.18502/ijph.v50i1.5082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Background We attempt to identify specific differentially methylated and expressed genes in people with longevity family history, it will contribute to discover significant features about human longevity. Methods A prevalence study was conducted during October 2017 to January 2019 in Bama County of Guangxi, China and individuals were recruited and grouped into longevity family (n=60) and non-longevity family (n=60) to identify differentially methylated genes (DMGs). The expression profile dataset GSE16717 was downloaded from the GEO database in which individuals were divided into 3 groups, namely longevity (n=50), longevity offspring (n=50) and control (n=50) for identifying differentially expressed genes (DEGs). It was considered significantly different when P or adjusted P≤0.05. Results In total, 117 longevity-related hypermethylated genes enriched in interleukin secretion/production regulation, chemokine signaling pathway and natural killer cell-mediated cytotoxicity. Another 296 significant key longevity-related DEGs primarily involved in protein binding, nucleus, cytoplasm, T cell receptor signaling pathway and Metabolic pathway, H19 and PFKFB4 were found to be both methylated and downregulated in people with longevity family history. Conclusion Human longevity-specific genes involve in many immunity regulations and cellular immunity pathways, H19 and PFKFB4 show hypermethylated and suppressed status in people with longevity family history and might serve as longevity candidate genes.
Collapse
Affiliation(s)
- Chunhong Li
- Department of Environmental Health, School of Public Health, Guangxi Medical University, Nanning, China
| | - Qingqing Nong
- Department of Environmental Health, School of Public Health, Guangxi Medical University, Nanning, China
| | - Bin Guan
- Department of Environmental Health, School of Public Health, Guangxi Medical University, Nanning, China
| | - Haoyu He
- Department of Environmental Health, School of Public Health, Guangxi Medical University, Nanning, China
| | - Zhiyong Zhang
- Department of Environmental Health, School of Public Health, Guangxi Medical University, Nanning, China.,Department of Environmental Health, School of Public Health, Guilin medical University, Guilin, China
| |
Collapse
|
24
|
Ustunel S, Prévôt ME, Clements RJ, Hegmann E. Cradle-to-cradle: designing biomaterials to fit as truly biomimetic cell scaffolds– a review. LIQUID CRYSTALS TODAY 2020. [DOI: 10.1080/1358314x.2020.1855919] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Senay Ustunel
- Materials Science Graduate Program, Kent State University, Kent, OH, USA
- Advanced Materials and Liquid Crystal Institute, Kent State University, Kent, OH, USA
| | - Marianne E. Prévôt
- Advanced Materials and Liquid Crystal Institute, Kent State University, Kent, OH, USA
| | - Robert J. Clements
- Department of Biological Sciences, Kent State University, Kent, OH, USA
- Biomedical Sciences Program, Kent State University, Kent, OH, USA
- Brain Health Research Institute, Kent State University, Kent, OH, USA
| | - Elda Hegmann
- Materials Science Graduate Program, Kent State University, Kent, OH, USA
- Advanced Materials and Liquid Crystal Institute, Kent State University, Kent, OH, USA
- Department of Biological Sciences, Kent State University, Kent, OH, USA
- Biomedical Sciences Program, Kent State University, Kent, OH, USA
- Brain Health Research Institute, Kent State University, Kent, OH, USA
| |
Collapse
|
25
|
Pharmacokinetic and Pharmacodynamic Modeling to Optimize the Dose of Vestronidase Alfa, an Enzyme Replacement Therapy for Treatment of Patients with Mucopolysaccharidosis Type VII: Results from Three Trials. Clin Pharmacokinet 2020; 58:673-683. [PMID: 30467742 PMCID: PMC6451706 DOI: 10.1007/s40262-018-0721-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Introduction Mucopolysaccharidosis type VII (MPS VII, Sly Syndrome) is a progressive, debilitating, ultra-rare lysosomal storage disorder caused by the deficiency of β-glucuronidase (GUS), an enzyme required for breakdown of glycosaminoglycans (GAGs). Vestronidase alfa, a recombinant human GUS, is an enzyme replacement therapy approved in the US and EU for the treatment of MPS VII. Methods The pharmacokinetics (PK) and pharmacodynamics (PD) of vestronidase alfa were evaluated in 23 adult and pediatric subjects with MPS VII enrolled in phase I–III clinical trials to optimize the clinical dosing regimen of vestronidase alfa. The serum concentration-time profiles were adequately described by a two-compartment population PK model incorporating subjects’ body weight as the only significant covariate. Results Model-based simulations predicted a substantially decreased time duration of serum exposures exceeding the level of Kuptake (the in vitro determined vestronidase alfa concentration corresponding to 50% maximum rate of cellular uptake) for 4 or 8 mg/kg once every 4 weeks dosing, compared with 4 mg/kg once every other week (QOW) dosing by intravenous infusion, suggesting that given the same total monthly dose, the QOW dosing frequency should result in more efficient delivery to the GUS-deficient tissue cells, and therefore superior treatment efficacy. A standard inhibitory maximal effect model reasonably explained the observed pharmacological PD responses of reduction in urinary GAGs from pretreatment baseline, which appeared to have reached the plateau of maximal effect at the 4 mg/kg QOW dose. Conclusion The modeling results, together with the clinical evidence of safety and efficacy, supported the recommended 4 mg/kg QOW dosing regimen of vestronidase alfa for pediatric and adult patients with MPS VII. Clinical Trial Registration NCT01856218, NCT02418455, NCT02230566. Electronic supplementary material The online version of this article (10.1007/s40262-018-0721-y) contains supplementary material, which is available to authorized users.
Collapse
|
26
|
Humphreys SC, Thayer MB, Campbell J, Chen WLK, Adams D, Lade JM, Rock BM. Emerging siRNA Design Principles and Consequences for Biotransformation and Disposition in Drug Development. J Med Chem 2020; 63:6407-6422. [PMID: 32352779 DOI: 10.1021/acs.jmedchem.9b01839] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
After two decades teetering at the intersection of laboratory tool and therapeutic reality, with two siRNA drugs now clinically approved, this modality has finally come into fruition. Consistent with other emerging modalities, initial proof-of-concept efforts concentrated on coupling pharmacologic efficacy with desirable safety profiles. Consequently, thorough investigations of siRNA absorption, distribution, metabolism, and excretion (ADME) properties are lacking. Advancing ADME knowledge will aid establishment of in vitro-in vivo correlations and pharmacokinetic-pharmacodynamic relationships to optimize candidate selection through discovery and translation. Here, we outline the emerging siRNA design principles and discuss the consequences for siRNA disposition and biotransformation. We propose a conceptual framework for siRNA ADME evaluation, contextualizing the site of biotransformation product formation with PK-PD modulation, and end with a discussion around safety and regulatory considerations and future directions for this modality.
Collapse
Affiliation(s)
- Sara C Humphreys
- Pharmacokinetics and Drug Metabolism Department, Amgen Research, 1120 Veterans Boulevard, South San Francisco, California 94080, United States
| | - Mai B Thayer
- Pharmacokinetics and Drug Metabolism Department, Amgen Research, 1120 Veterans Boulevard, South San Francisco, California 94080, United States
| | - Jabbar Campbell
- Neuroscience Department, Amgen Research, 360 Binney Street, Cambridge, Massachusetts 02141, United States
| | - Wen Li Kelly Chen
- Comparative Biology and Safety Sciences Department, Amgen Research, 360 Binney Street, Cambridge, Massachusetts 02141, United States
| | - Dan Adams
- Comparative Biology and Safety Sciences Department, Amgen Research, 360 Binney Street, Cambridge, Massachusetts 02141, United States
| | - Julie M Lade
- Pharmacokinetics and Drug Metabolism Department, Amgen Research, 1120 Veterans Boulevard, South San Francisco, California 94080, United States
| | - Brooke M Rock
- Pharmacokinetics and Drug Metabolism Department, Amgen Research, 1120 Veterans Boulevard, South San Francisco, California 94080, United States
| |
Collapse
|
27
|
Andrawus M, Sharvit L, Shekhidem HA, Roichman A, Cohen HY, Atzmon G. The effects of environmental stressors on candidate aging associated genes. Exp Gerontol 2020; 137:110952. [PMID: 32344118 DOI: 10.1016/j.exger.2020.110952] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 03/29/2020] [Accepted: 04/09/2020] [Indexed: 01/15/2023]
Abstract
BACKGROUND Aging is defined as a biological and physical complex process that is characterized by the increase in susceptibility to diseases and eventually death. Aging may occur at different rates between and within species, especially or (it varies) among the long-lived ones. Here, we ask whether this diversity (e.g. aging phenotype) stems from genetic or environmental factors or as a combination between the two (epigenetics). Epigenetics play a central role in controlling changes in gene expression during aging. DNA methylation is the most abundant epigenetic modification among vertebrates and is essential to mammalian development. MATERIALS AND METHODS In this study, we utilized the HELPtag assay to identify five candidate genes that were significantly hyper- or hypo-methylated across four different age groups in mice. The candidate genes were annotated using ensemble and their expression was further tested in vitro using the murine RAW 264.7 cell line to examine the effect of three environmental stressors (UV radiation, Hypoxia and fasting) on their expression. RNA was extracted at different time points followed by cDNA synthesis. Changes in gene expression were evaluated using qRT-PCR. RESULTS We show that fasting and UV radiation reduced the viability of RAW264.7 cells. We also found a significant change in three candidate genes' expression levels during fasting (TOP2B, RNF13 and MRPL4). Furthermore, we found a significant change in the four candidate genes' expression levels following UVC treatment (TOP2B, RNF13, PKNOX1 and CREB5) and yet no changes were recorded in hypoxic conditions. CONCLUSION Our results suggest that the model we used was a fitting model for the assessment of environmental stressors on candidate gene expression. In addition, we established a cellular response to the environment via changes in gene expression.
Collapse
Affiliation(s)
- Mariana Andrawus
- Department of Human Biology, University of Haifa, Haifa 3498838, Israel
| | - Lital Sharvit
- Department of Human Biology, University of Haifa, Haifa 3498838, Israel
| | | | - Asael Roichman
- Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Haim Y Cohen
- Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Gil Atzmon
- Department of Human Biology, University of Haifa, Haifa 3498838, Israel.
| |
Collapse
|
28
|
Capuco AV, Choudhary RK. Symposium review: Determinants of milk production: Understanding population dynamics in the bovine mammary epithelium. J Dairy Sci 2020; 103:2928-2940. [PMID: 31704023 DOI: 10.3168/jds.2019-17241] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 09/23/2019] [Indexed: 01/11/2025]
Abstract
The mammary gland undergoes distinct periods of growth, development, and secretory activity. During bovine lactation, a gradual decrease in the number of mammary epithelial cells largely accounts for the decline in milk production with advancing lactation. The net decline in cell number (approx. 50%) is due to cell death but is simultaneously accompanied by cell renewal. Although the rate of cell proliferation is slow, by the end of lactation most cells in the gland were formed after calving. Typically milking is terminated when cows are in the final 2 mo of pregnancy. This causes regenerative involution, wherein extensive cell replacement and mammary growth occurs. We hypothesized that replacement of senescent secretory cells and progenitor cells during the dry period increases milk yield in the next lactation. Analysis of global gene expression revealed networks and canonical pathways during regenerative involution that support cell turnover and mammary growth, and reflect oxidative stress, mitochondrial dysfunction, and endoplasmic reticulum (ER) stress. Immune responses consistent with influx of neutrophils, macrophages, and lymphocytes, and processes that support mammary differentiation and lactogenesis were also evident. Data also suggest that replication of stem and progenitor cells occurs during the dry period. Relying on long-term retention of bromodeoxyuridine-labeled DNA, we identified putative bovine mammary stem cells. These label-retaining epithelial cells (LREC) are in low abundance within mammary epithelium (<1%), predominantly estrogen receptor-negative, and localized in a basal or suprabasal layer of the epithelium. Analyses of gene expression in laser-microdissected LREC are consistent with the concept that LREC represent stem cells and progenitor cells, which differ in properties and location within the epithelial layer. We identified potential markers for these cells and have increased their number by infusing xanthosine through the teat canal of prepubertal heifers. Altering population dynamics of mammary stem and progenitor cells during the mammary cycle may be a means to increase efficiency of milk production.
Collapse
Affiliation(s)
- Anthony V Capuco
- Animal Genomics and Improvement Laboratory, USDA-Agricultural Research Service, Beltsville, MD 20705.
| | - Ratan K Choudhary
- College of Animal Biotechnology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, Punjab 141004, India
| |
Collapse
|
29
|
Bakula D, Ablasser A, Aguzzi A, Antebi A, Barzilai N, Bittner MI, Jensen MB, Calkhoven CF, Chen D, de Grey AD, Feige JN, Georgievskaya A, Gladyshev VN, Golato T, Gudkov AV, Hoppe T, Kaeberlein M, Katajisto P, Kennedy BK, Lal U, Martin-Villalba A, Moskalev AA, Ozerov I, Petr MA, Reason, Rubinsztein DC, Tyshkovskiy A, Vanhaelen Q, Zhavoronkov A, Scheibye-Knudsen M. Latest advances in aging research and drug discovery. Aging (Albany NY) 2019; 11:9971-9981. [PMID: 31770722 PMCID: PMC6914421 DOI: 10.18632/aging.102487] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 11/09/2019] [Indexed: 12/19/2022]
Abstract
An increasing aging population poses a significant challenge to societies worldwide. A better understanding of the molecular, cellular, organ, tissue, physiological, psychological, and even sociological changes that occur with aging is needed in order to treat age-associated diseases. The field of aging research is rapidly expanding with multiple advances transpiring in many previously disconnected areas. Several major pharmaceutical, biotechnology, and consumer companies made aging research a priority and are building internal expertise, integrating aging research into traditional business models and exploring new go-to-market strategies. Many of these efforts are spearheaded by the latest advances in artificial intelligence, namely deep learning, including generative and reinforcement learning. To facilitate these trends, the Center for Healthy Aging at the University of Copenhagen and Insilico Medicine are building a community of Key Opinion Leaders (KOLs) in these areas and launched the annual conference series titled "Aging Research and Drug Discovery (ARDD)" held in the capital of the pharmaceutical industry, Basel, Switzerland (www.agingpharma.org). This ARDD collection contains summaries from the 6th annual meeting that explored aging mechanisms and new interventions in age-associated diseases. The 7th annual ARDD exhibition will transpire 2nd-4th of September, 2020, in Basel.
Collapse
Affiliation(s)
- Daniela Bakula
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Andrea Ablasser
- Global Health Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Adriano Aguzzi
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland
| | - Adam Antebi
- Max Planck Institute for Biology of Ageing, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Nir Barzilai
- Department of Medicine, Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | - Cornelis F. Calkhoven
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, AD Groningen, The Netherlands
| | - Danica Chen
- Program in Metabolic Biology, Nutritional Sciences and Toxicology, University of California, Berkeley, CA 94720, USA
| | | | - Jerome N. Feige
- Nestlé Research, EPFL Innovation Park, Lausanne, Switzerland
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | | | - Vadim N. Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | - Andrei V. Gudkov
- Roswell Park Comprehensive Cancer Center and Genome Protection, Inc., Buffalo, NY 14203, USA
| | - Thorsten Hoppe
- Institute for Genetics and CECAD Research Center, University of Cologne, Cologne, Germany
| | - Matt Kaeberlein
- Department of Pathology, School of Medicine, University of Washington, Seattle, WA 98195, USA
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Pekka Katajisto
- Department of Biosciences and Nutrition, Center for Innovative Medicine, Karolinska Institutet, Huddinge, Sweden
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Brian K. Kennedy
- Departments of Biochemistry and Physiology, Yong Loo Lin School of Medicine, National University Singapore, Singapore
- Centre for Healthy Ageing, National University Healthy System, Singapore
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Unmesh Lal
- Frost and Sullivan, Frankfurt am Main, Germany
| | | | - Alexey A. Moskalev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Institute of Biology of Komi Science Center of Ural Branch of RAS, Syktyvkar, Russia
- Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Ivan Ozerov
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Rockville, MD 20850, USA
| | - Michael A. Petr
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Reason
- Repair Biotechnologies, Inc., Syracuse, NY 13210, USA
| | - David C. Rubinsztein
- Department of Medical Genetics, Cambridge Institute for Medical Research, The Keith Peters Building, Cambridge CB2 0XY, UK
- UK Dementia Research Institute, The Keith Peters Building, Cambridge Biomedical Campus, Cambridge, UK
| | - 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, Russia
| | - Quentin Vanhaelen
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Rockville, MD 20850, USA
| | - Alex Zhavoronkov
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Rockville, MD 20850, USA
| | - Morten Scheibye-Knudsen
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
30
|
Morris BJ, Willcox BJ, Donlon TA. Genetic and epigenetic regulation of human aging and longevity. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1718-1744. [PMID: 31109447 PMCID: PMC7295568 DOI: 10.1016/j.bbadis.2018.08.039] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 08/02/2018] [Accepted: 08/28/2018] [Indexed: 02/06/2023]
Abstract
Here we summarize the latest data on genetic and epigenetic contributions to human aging and longevity. Whereas environmental and lifestyle factors are important at younger ages, the contribution of genetics appears more important in reaching extreme old age. Genome-wide studies have implicated ~57 gene loci in lifespan. Epigenomic changes during aging profoundly affect cellular function and stress resistance. Dysregulation of transcriptional and chromatin networks is likely a crucial component of aging. Large-scale bioinformatic analyses have revealed involvement of numerous interaction networks. As the young well-differentiated cell replicates into eventual senescence there is drift in the highly regulated chromatin marks towards an entropic middle-ground between repressed and active, such that genes that were previously inactive "leak". There is a breakdown in chromatin connectivity such that topologically associated domains and their insulators weaken, and well-defined blocks of constitutive heterochromatin give way to generalized, senescence-associated heterochromatin, foci. Together, these phenomena contribute to aging.
Collapse
Affiliation(s)
- Brian J Morris
- Basic & Clinical Genomics Laboratory, School of Medical Sciences and Bosch Institute, University of Sydney, New South Wales 2006, Australia; Honolulu Heart Program (HHP)/Honolulu-Asia Aging Study (HAAS), Department of Research, Kuakini Medical Center, Honolulu, HI 96817, United States; Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Kuakini Medical Center Campus, Honolulu, HI 96813, United States.
| | - Bradley J Willcox
- Honolulu Heart Program (HHP)/Honolulu-Asia Aging Study (HAAS), Department of Research, Kuakini Medical Center, Honolulu, HI 96817, United States; Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Kuakini Medical Center Campus, Honolulu, HI 96813, United States.
| | - Timothy A Donlon
- Honolulu Heart Program (HHP)/Honolulu-Asia Aging Study (HAAS), Department of Research, Kuakini Medical Center, Honolulu, HI 96817, United States; Departments of Cell & Molecular Biology and Pathology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, United States.
| |
Collapse
|
31
|
Zhang W, Feng Y, Guo Q, Guo W, Xu H, Li X, Yi F, Guan Y, Geng N, Wang P, Cao L, O'Rourke BP, Jo J, Kwon J, Wang R, Song X, Lee IH, Cao L. SIRT1 modulates cell cycle progression by regulating CHK2 acetylation-phosphorylation. Cell Death Differ 2019; 27:482-496. [PMID: 31209362 PMCID: PMC7206007 DOI: 10.1038/s41418-019-0369-7] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 04/26/2019] [Accepted: 05/27/2019] [Indexed: 12/21/2022] Open
Abstract
Both the stress-response protein, SIRT1, and the cell cycle checkpoint kinase, CHK2, play critical roles in aging and cancer via the modulation of cellular homeostasis and the maintenance of genomic integrity. However, the underlying mechanism linking the two pathways remains elusive. Here, we show that SIRT1 functions as a modifier of CHK2 in cell cycle control. Specifically, SIRT1 interacts with CHK2 and deacetylates it at lysine 520 residue, which suppresses CHK2 phosphorylation, dimerization, and thus activation. SIRT1 depletion induces CHK2 hyperactivation-mediated cell cycle arrest and subsequent cell death. In vivo, genetic deletion of Chk2 rescues the neonatal lethality of Sirt1−/− mice, consistent with the role of SIRT1 in preventing CHK2 hyperactivation. Together, these results suggest that CHK2 mediates the function of SIRT1 in cell cycle progression, and may provide new insights into modulating cellular homeostasis and maintaining genomic integrity in the prevention of aging and cancer.
Collapse
Affiliation(s)
- Wenyu Zhang
- Institute of Translational Medicine, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China
| | - Yanling Feng
- Institute of Translational Medicine, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China
| | - Qiqiang Guo
- Institute of Translational Medicine, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China
| | - Wendong Guo
- Institute of Translational Medicine, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China
| | - Hongde Xu
- Institute of Translational Medicine, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China
| | - Xiaoman Li
- Institute of Translational Medicine, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China
| | - Fei Yi
- Institute of Translational Medicine, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China
| | - Yi Guan
- Institute of Translational Medicine, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China
| | - Nanxi Geng
- Department of Cell Biology, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China
| | - Pingyuan Wang
- Center for Molecular Medicine, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Longyue Cao
- Department of Medicine (Cardiology), Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Brian P O'Rourke
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Juhyeon Jo
- Department of Life Science, College of Natural Science Office #106, Science building C, Ewha Womans University 52, Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, South Korea
| | - Jiyun Kwon
- Department of Life Science, College of Natural Science Office #106, Science building C, Ewha Womans University 52, Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, South Korea
| | - Ruihong Wang
- Faculty of Health Science, University of Macau, Macau, China
| | - Xiaoyu Song
- Institute of Translational Medicine, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China.
| | - In Hye Lee
- Department of Life Science, College of Natural Science Office #106, Science building C, Ewha Womans University 52, Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, South Korea.
| | - Liu Cao
- Institute of Translational Medicine, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China.
| |
Collapse
|
32
|
Teimuri S, Hosseini A, Rezaenasab A, Ghaedi K, Ghoveud E, Etemadifar M, Nasr-Esfahani MH, Megraw TL. Integrative Analysis of lncRNAs in Th17 Cell Lineage to Discover New Potential Biomarkers and Therapeutic Targets in Autoimmune Diseases. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 12:393-404. [PMID: 30195777 PMCID: PMC6128809 DOI: 10.1016/j.omtn.2018.05.022] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Revised: 05/24/2018] [Accepted: 05/24/2018] [Indexed: 11/27/2022]
Abstract
Th17 cells play a critical role in the pathogenesis of autoimmune diseases, including multiple sclerosis, rheumatoid arthritis, systemic lupus erythematosus, Sjogren's syndrome, and inflammatory bowel disease. Despite the extensive investigation into this T cell lineage, little is understood regarding the role of Th17 lineage-specific lncRNAs (long non-coding RNAs) > 200 nt. lncRNAs may influence disease through a variety of mechanisms; their expression could be regulated by SNPs. lncRNAs can also affect the expression of neighboring genes or complementary miRNAs, and their expression may have lineage-specific patterns. In the system biology study presented here, the effective lncRNAs from different criteria were predicted for each autoimmune disease, and we then evaluated their expression levels in 50 MS patients compared to 25 controls using qRT-PCR. We identified changes in the expression levels of AL450992.2, AC009948.5, and RP11-98D18.3 as potential peripheral blood mononuclear cell (PBMC) biomarkers for MS among our studied lncRNAs in which co-expression analysis of AL450992.2 had the most AUCs, and the relationship to RORC was also assessed. We propose that the recurrently deregulated lncRNAs identified in this report could provide a valuable resource for studies aimed at delineating the relationship between functional lncRNAs and autoimmune disorders.
Collapse
Affiliation(s)
- Shohreh Teimuri
- Division of Cellular and Molecular Biology, Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran
| | - Aref Hosseini
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Ahmad Rezaenasab
- Division of Cellular and Molecular Biology, Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran
| | - Kamran Ghaedi
- Division of Cellular and Molecular Biology, Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran; Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.
| | - Elahe Ghoveud
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Masoud Etemadifar
- Department of Neurology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Hossein Nasr-Esfahani
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.
| | - Timothy L Megraw
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, USA.
| |
Collapse
|
33
|
Garcia S, Nissanka N, Mareco EA, Rossi S, Peralta S, Diaz F, Rotundo RL, Carvalho RF, Moraes CT. Overexpression of PGC-1α in aging muscle enhances a subset of young-like molecular patterns. Aging Cell 2018; 17:e12707. [PMID: 29427317 PMCID: PMC5847875 DOI: 10.1111/acel.12707] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/31/2017] [Indexed: 12/31/2022] Open
Abstract
PGC-1α is a transcriptional co-activator known as the master regulator of mitochondrial biogenesis. Its control of metabolism has been suggested to exert critical influence in the aging process. We have aged mice overexpressing PGC-1α in skeletal muscle to determine whether the transcriptional changes reflected a pattern of expression observed in younger muscle. Analyses of muscle proteins showed that Pax7 and several autophagy markers were increased. In general, the steady-state levels of several muscle proteins resembled that of muscle from young mice. Age-related mtDNA deletion levels were not increased by the PGC-1α-associated increase in mitochondrial biogenesis. Accordingly, age-related changes in the neuromuscular junction were minimized by PGC-1α overexpression. RNA-Seq showed that several genes overexpressed in the aged PGC-1α transgenic are expressed at higher levels in young when compared to aged skeletal muscle. As expected, there was increased expression of genes associated with energy metabolism but also of pathways associated with muscle integrity and regeneration. We also found that PGC-1α overexpression had a mild but significant effect on longevity. Taken together, overexpression of PGC-1α in aged muscle led to molecular changes that resemble the patterns observed in skeletal muscle from younger mice.
Collapse
Affiliation(s)
- Sofia Garcia
- Department of NeurologyUniversity of Miami Miller School of MedicineMiamiFLUSA
| | - Nadee Nissanka
- Neuroscience Graduate ProgramUniversity of Miami Miller School of MedicineMiamiFLUSA
| | - Edson A. Mareco
- Graduate Program in Environment and Regional DevelopmentUniversity of Western São PauloPresidente PrudenteBrazil
| | - Susana Rossi
- Department of Cell BiologyUniversity of Miami Miller School of MedicineMiamiFLUSA
| | - Susana Peralta
- Department of NeurologyUniversity of Miami Miller School of MedicineMiamiFLUSA
| | - Francisca Diaz
- Department of NeurologyUniversity of Miami Miller School of MedicineMiamiFLUSA
| | - Richard L. Rotundo
- Neuroscience Graduate ProgramUniversity of Miami Miller School of MedicineMiamiFLUSA
- Department of Cell BiologyUniversity of Miami Miller School of MedicineMiamiFLUSA
| | - Robson F. Carvalho
- Institute of BiosciencesSão Paulo State University (UNESP)BotucatuBrazil
| | - Carlos T. Moraes
- Department of NeurologyUniversity of Miami Miller School of MedicineMiamiFLUSA
- Neuroscience Graduate ProgramUniversity of Miami Miller School of MedicineMiamiFLUSA
- Department of Cell BiologyUniversity of Miami Miller School of MedicineMiamiFLUSA
| |
Collapse
|
34
|
Abstract
Ageing leads to dramatic changes in the physiology of many different tissues resulting in a spectrum of pathology. Nonetheless, many lines of evidence suggest that ageing is driven by highly conserved cell intrinsic processes, and a set of unifying hallmarks of ageing has been defined. Here, we survey reports of age-linked changes in basal gene expression across eukaryotes from yeast to human and identify six gene expression hallmarks of cellular ageing: downregulation of genes encoding mitochondrial proteins; downregulation of the protein synthesis machinery; dysregulation of immune system genes; reduced growth factor signalling; constitutive responses to stress and DNA damage; dysregulation of gene expression and mRNA processing. These encompass widely reported features of ageing such as increased senescence and inflammation, reduced electron transport chain activity and reduced ribosome synthesis, but also reveal a surprising lack of gene expression responses to known age-linked cellular stresses. We discuss how the existence of conserved transcriptomic hallmarks relates to genome-wide epigenetic differences underlying ageing clocks, and how the changing transcriptome results in proteomic alterations where data is available and to variations in cell physiology characteristic of ageing. Identification of gene expression events that occur during ageing across distant organisms should be informative as to conserved underlying mechanisms of ageing, and provide additional biomarkers to assess the effects of diet and other environmental factors on the rate of ageing.
Collapse
Affiliation(s)
- Stephen Frenk
- Department of Genetics, University of North Carolina, Chapel Hill, NC, 27599-3280, USA
| | | |
Collapse
|
35
|
Dealing with Confounders in Omics Analysis. Trends Biotechnol 2018; 36:488-498. [PMID: 29475622 DOI: 10.1016/j.tibtech.2018.01.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 01/28/2018] [Accepted: 01/29/2018] [Indexed: 01/05/2023]
Abstract
The Anna Karenina effect is a manifestation of the theory-practice gap that exists when theoretical statistics are applied on real-world data. In the course of analyzing biological data for differential features such as genes or proteins, it derives from the situation where the null hypothesis is rejected for extraneous reasons (or confounders), rather than because the alternative hypothesis is relevant to the disease phenotype. The mechanics of applying statistical tests therefore must address and resolve confounders. It is inadequate to simply rely on manipulating the P-value. We discuss three mechanistic elements (hypothesis statement construction, null distribution appropriateness, and test-statistic construction) and suggest how they can be designed to foil the Anna Karenina effect to select phenotypically relevant biological features.
Collapse
|
36
|
Keyes BE, Fuchs E. Stem cells: Aging and transcriptional fingerprints. J Cell Biol 2017; 217:79-92. [PMID: 29070608 PMCID: PMC5748991 DOI: 10.1083/jcb.201708099] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 10/06/2017] [Accepted: 10/10/2017] [Indexed: 12/31/2022] Open
Abstract
Keyes and Fuchs discuss the decline in stem cell renewal and function with aging and the ensuing consequences on tissue homeostasis and regeneration. Stem cells are imbued with unique qualities. They have the capacity to propagate themselves through symmetric divisions and to divide asymmetrically to engender new cells that can progress to differentiate into tissue-specific, terminal cell types. Armed with these qualities, stem cells in adult tissues are tasked with replacing decaying cells and regenerating tissue after injury to maintain optimal tissue function. With increasing age, stem cell functional abilities decline, resulting in reduced organ function and delays in tissue repair. Here, we review the effect of aging in five well-studied adult murine stem cell populations and explore age-related declines in stem cell function and their consequences for stem cell self-renewal, tissue homeostasis, and regeneration. Finally, we examine transcriptional changes that have been documented in aged stem cell populations and discuss new questions and future directions that this collection of data has uncovered.
Collapse
Affiliation(s)
| | - Elaine Fuchs
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, Howard Hughes Medical Institute, The Rockefeller University, New York, NY
| |
Collapse
|
37
|
Pappas K, Xu J, Zairis S, Resnick-Silverman L, Abate F, Steinbach N, Ozturk S, Saal LH, Su T, Cheung P, Schmidt H, Aaronson S, Hibshoosh H, Manfredi J, Rabadan R, Parsons R. p53 Maintains Baseline Expression of Multiple Tumor Suppressor Genes. Mol Cancer Res 2017; 15:1051-1062. [PMID: 28483946 DOI: 10.1158/1541-7786.mcr-17-0089] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 04/21/2017] [Accepted: 05/02/2017] [Indexed: 02/07/2023]
Abstract
TP53 is the most commonly mutated tumor suppressor gene and its mutation drives tumorigenesis. Using ChIP-seq for p53 in the absence of acute cell stress, we found that wild-type but not mutant p53 binds and activates numerous tumor suppressor genes, including PTEN, STK11(LKB1), miR-34a, KDM6A(UTX), FOXO1, PHLDA3, and TNFRSF10B through consensus binding sites in enhancers and promoters. Depletion of p53 reduced expression of these target genes, and analysis across 18 tumor types showed that mutation of TP53 associated with reduced expression of many of these genes. Regarding PTEN, p53 activated expression of a luciferase reporter gene containing the p53-consensus site in the PTEN enhancer, and homozygous deletion of this region in cells decreased PTEN expression and increased growth and transformation. These findings show that p53 maintains expression of a team of tumor suppressor genes that may together with the stress-induced targets mediate the ability of p53 to suppress cancer development. p53 mutations selected during tumor initiation and progression, thus, inactivate multiple tumor suppressor genes in parallel, which could account for the high frequency of p53 mutations in cancer.Implications: In this study, we investigate the activities of p53 under normal low-stress conditions and discover that p53 is capable of maintaining the expression of a group of important tumor suppressor genes at baseline, many of which are haploinsufficient, which could contribute to p53-mediated tumor suppression. Mol Cancer Res; 15(8); 1051-62. ©2017 AACR.
Collapse
Affiliation(s)
- Kyrie Pappas
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Pharmacology, Columbia University Medical Center, New York, New York
| | - Jia Xu
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Sakellarios Zairis
- Department of Systems Biology, Columbia University Medical Center, New York, New York
| | - Lois Resnick-Silverman
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Francesco Abate
- Department of Systems Biology, Columbia University Medical Center, New York, New York.,Department of Biomedical Informatics, Columbia University Medical Center, New York, New York
| | - Nicole Steinbach
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Graduate School of Arts and Sciences, Columbia University Medical Center, New York, New York
| | - Sait Ozturk
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Lao H Saal
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden.,Lund University Cancer Center, Lund, Sweden.,CREATE Health Strategic Center for Translational Cancer Research, Lund University, Lund, Sweden
| | - Tao Su
- Department of Pathology, Columbia University Medical Center, New York, New York
| | - Pamela Cheung
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Hank Schmidt
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Dubin Breast Center, The Mount Sinai Hospital, New York, New York.,Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Stuart Aaronson
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Hanina Hibshoosh
- Department of Pathology, Columbia University Medical Center, New York, New York
| | - James Manfredi
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Raul Rabadan
- Department of Systems Biology, Columbia University Medical Center, New York, New York.,Department of Biomedical Informatics, Columbia University Medical Center, New York, New York
| | - Ramon Parsons
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York. .,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
38
|
Seim I, Crisp G, Shah ET, Jeffery PL, Chopin LK. Abundant ghrelin gene expression by monocytes: Putative implications for fat accumulation and obesity. ACTA ACUST UNITED AC 2017. [DOI: 10.1016/j.obmed.2016.12.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
39
|
Abstract
Circular RNAs (circRNAs) are a newly appreciated class of RNAs expressed across diverse phyla. These enigmatic transcripts are most commonly generated by back-splicing events from exons of protein-coding genes. This results in highly stable RNAs due to the lack of free 5′ and 3′ ends. CircRNAs are enriched in neural tissues, suggesting that they might have neural functions. Here, we sought to determine whether circRNA accumulation occurs during aging in mice. Total RNA-seq profiling of young (1 month old) and aged (22 month old) cortex, hippocampus and heart samples was performed. This led to the confident detection of 6,791 distinct circRNAs across these samples, including 675 novel circRNAs. Analysis uncovered a strong bias for circRNA upregulation during aging in neural tissues. These age-accumulation trends were verified for individual circRNAs by RT-qPCR and Northern analysis. In contrast, comparison of aged versus young hearts failed to reveal a global trend for circRNA upregulation. Age-accumulation of circRNAs in brain tissues was found to be largely independent from linear RNA expression of host genes. These findings suggest that circRNAs might play biological roles relevant to the aging nervous system.
Collapse
|