1
|
Castillo-Mariqueo L, Alveal-Mellado D, Giménez-Llort L. Phenotypical, Behavioral, and Systemic Hallmarks in End-Point Mouse Scenarios. Animals (Basel) 2025; 15:521. [PMID: 40003003 PMCID: PMC11851987 DOI: 10.3390/ani15040521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 01/31/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
The state of frailty is a clinical-biological syndrome that affects the older population with a higher risk of functional dependence. Animal models can provide a tool to study this complex scenario. In the present work, we analyzed the physical and behavioral hallmarks of end-point status in 16-month-old mice (C57BL/6J) according to animal welfare regulations compared to age-matched counterparts with normal aging. A group of 6-month-old mice was added to control for age bias. First, we identified 'structural kyphosis' (visible and unmodifiable deformation in locomotion) correlated with piloerection as the hallmarks of the physical frailty phenotype compared to the 'postural kyphosis' (adjustment to counteract increased visceral volume but attenuated during locomotion) of old mice with normal aging. Alopecia (barbering) was presented in both old groups. Normal levels of exploratory activity in the corner test for neophobia and triceps surae muscle weight but an increased latency of rearing indicated the poorest emotional phenotype, with a possible contribution of structural kyphosis. The presence of hepatomegaly and splenomegaly counteracted the significant WAT loss commonly associated with end-of-life traits, which should have a normal body weight but preserved muscle mass.
Collapse
Affiliation(s)
- Lidia Castillo-Mariqueo
- Institut de Neurociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193 Barcelona, Spain; (L.C.-M.); (D.A.-M.)
- Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193 Barcelona, Spain
- Departamento de Procesos Terapeúticos, Facultad de Ciencias de la Salud, Universidad Católica de Temuco, Temuco 4780000, Chile
| | - Daniel Alveal-Mellado
- Institut de Neurociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193 Barcelona, Spain; (L.C.-M.); (D.A.-M.)
- Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193 Barcelona, Spain
| | - Lydia Giménez-Llort
- Institut de Neurociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193 Barcelona, Spain; (L.C.-M.); (D.A.-M.)
- Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193 Barcelona, Spain
| |
Collapse
|
2
|
Zhao JV, Yao M, Liu Z. Using genetics and proteomics data to identify proteins causally related to COVID-19, healthspan and lifespan: a Mendelian randomization study. Aging (Albany NY) 2024; 16:6384-6416. [PMID: 38575325 PMCID: PMC11042960 DOI: 10.18632/aging.205711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 01/24/2024] [Indexed: 04/06/2024]
Abstract
BACKGROUND COVID-19 pandemic poses a heavy burden on public health and accounts for substantial mortality and morbidity. Proteins are building blocks of life, but specific proteins causally related to COVID-19, healthspan and lifespan have not been systematically examined. METHODS We conducted a Mendelian randomization study to assess the effects of 1,361 plasma proteins on COVID-19, healthspan and lifespan, using large GWAS of severe COVID-19 (up to 13,769 cases and 1,072,442 controls), COVID-19 hospitalization (32,519 cases and 2,062,805 controls) and SARS-COV2 infection (122,616 cases and 2,475,240 controls), healthspan (n = 300,477) and parental lifespan (~0.8 million of European ancestry). RESULTS We identified 35, 43, and 63 proteins for severe COVID, COVID-19 hospitalization, and SARS-COV2 infection, and 4, 32, and 19 proteins for healthspan, father's attained age, and mother's attained age. In addition to some proteins reported previously, such as SFTPD related to severe COVID-19, we identified novel proteins involved in inflammation and immunity (such as ICAM-2 and ICAM-5 which affect COVID-19 risk, CXCL9, HLA-DRA and LILRB4 for healthspan and lifespan), apoptosis (such as FGFR2 and ERBB4 which affect COVID-19 risk and FOXO3 which affect lifespan) and metabolism (such as PCSK9 which lowers lifespan). We found 2, 2 and 3 proteins shared between COVID-19 and healthspan/lifespan, such as CXADR and LEFTY2, shared between severe COVID-19 and healthspan/lifespan. Three proteins affecting COVID-19 and seven proteins affecting healthspan/lifespan are targeted by existing drugs. CONCLUSIONS Our study provided novel insights into protein targets affecting COVID-19, healthspan and lifespan, with implications for developing new treatment and drug repurposing.
Collapse
Affiliation(s)
- Jie V. Zhao
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China
| | - Minhao Yao
- Department of Statistics and Actuarial Science, The University of Hong Kong, Hong Kong SAR, China
| | - Zhonghua Liu
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY 10032, USA
| |
Collapse
|
3
|
Omrani O, Krepelova A, Rasa SMM, Sirvinskas D, Lu J, Annunziata F, Garside G, Bajwa S, Reinhardt S, Adam L, Käppel S, Ducano N, Donna D, Ori A, Oliviero S, Rudolph KL, Neri F. IFNγ-Stat1 axis drives aging-associated loss of intestinal tissue homeostasis and regeneration. Nat Commun 2023; 14:6109. [PMID: 37777550 PMCID: PMC10542816 DOI: 10.1038/s41467-023-41683-y] [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: 10/28/2021] [Accepted: 09/14/2023] [Indexed: 10/02/2023] Open
Abstract
The influence of aging on intestinal stem cells and their niche can explain underlying causes for perturbation in their function observed during aging. Molecular mechanisms for such a decrease in the functionality of intestinal stem cells during aging remain largely undetermined. Using transcriptome-wide approaches, our study demonstrates that aging intestinal stem cells strongly upregulate antigen presenting pathway genes and over-express secretory lineage marker genes resulting in lineage skewed differentiation into the secretory lineage and strong upregulation of MHC class II antigens in the aged intestinal epithelium. Mechanistically, we identified an increase in proinflammatory cells in the lamina propria as the main source of elevated interferon gamma (IFNγ) in the aged intestine, that leads to the induction of Stat1 activity in intestinal stem cells thus priming the aberrant differentiation and elevated antigen presentation in epithelial cells. Of note, systemic inhibition of IFNγ-signaling completely reverses these aging phenotypes and reinstalls regenerative capacity of the aged intestinal epithelium.
Collapse
Affiliation(s)
- Omid Omrani
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Anna Krepelova
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
- Department of Life Sciences and Systems Biology, University of Turin, Torino, Italy
- Molecular Biotechnology Center, University of Turin, Torino, Italy
| | | | - Dovydas Sirvinskas
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Jing Lu
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | | | - George Garside
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Seerat Bajwa
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Susanne Reinhardt
- Dresden-concept Genome Center, c/o Center for Regenerative Therapies Dresden (CRTD), Dresden, Germany
| | - Lisa Adam
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Sandra Käppel
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Nadia Ducano
- Department of Life Sciences and Systems Biology, University of Turin, Torino, Italy
- Molecular Biotechnology Center, University of Turin, Torino, Italy
| | - Daniela Donna
- Department of Life Sciences and Systems Biology, University of Turin, Torino, Italy
- Molecular Biotechnology Center, University of Turin, Torino, Italy
| | - Alessandro Ori
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Salvatore Oliviero
- Department of Life Sciences and Systems Biology, University of Turin, Torino, Italy
- Molecular Biotechnology Center, University of Turin, Torino, Italy
| | | | - Francesco Neri
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany.
- Department of Life Sciences and Systems Biology, University of Turin, Torino, Italy.
- Molecular Biotechnology Center, University of Turin, Torino, Italy.
| |
Collapse
|
4
|
Perazza LR, Gower AC, Brown-Borg HM, Pajevic PD, Thompson LV. Protectin DX as a therapeutic strategy against frailty in mice. GeroScience 2023; 45:2601-2627. [PMID: 37059838 PMCID: PMC10651819 DOI: 10.1007/s11357-023-00789-3] [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: 01/03/2023] [Accepted: 03/31/2023] [Indexed: 04/16/2023] Open
Abstract
Frailty in aging is driven by the dysregulation of multiple biological pathways. Protectin DX (PDX) is a docosahexaenoic acid (DHA)-derived molecule that alleviates many chronic inflammatory disorders, but its potential effects on frailty remain unknown. Our goal is to identify age-related impairments in metabolic systems and to evaluate the therapeutic potential of PDX on frailty, physical performance, and health parameters. A set of 22-month-old C57BL/6 male and female mice were assigned to vehicle (Old) or PDX daily gavage treatment for 9 weeks, whereas 6-month-old (Adult) mice received only vehicle. Forelimb and hindlimb strength, endurance, voluntary wheel activity and walking speed determined physical performance and were combined with a frailty index score and body weight loss to determine frailty status. Our data shows that old vehicle-treated mice from both sexes had body weight loss paralleling visceromegaly, and Old females also had impaired insulin clearance as compared to the Adult group. Aging was associated with physical performance decline together with higher odds of frailty development. There was also age-driven mesangial expansion and glomerular hypertrophy as well as bone mineral density loss. All of the in vivo and in vitro impairments observed with aging co-occurred with upregulation of inflammatory pathways and Myc signaling as well as downregulation of genes related to adipogenesis and oxidative phosphorylation in liver. PDX attenuated the age-driven physical performance (strength, exhaustion, walking speed) decline, promoted robustness, prevented bone losses and partially reversed changes in hepatic expression of Myc targets and metabolic genes. In conclusion, our data provides evidence of the beneficial therapeutic effect of PDX against features of frailty in mice. Further studies are warranted to investigate the mechanisms of action and the potential for human translation.
Collapse
Affiliation(s)
- Laís R Perazza
- Department of Physical Therapy, Boston University, Boston, MA, USA.
| | - Adam C Gower
- Clinical and Translational Science Institute, Boston University, Boston, MA, USA
| | - Holly M Brown-Borg
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Paola Divieti Pajevic
- Department of Translational Dental Medicine, Goldman School of Dental Medicine, Boston University, Boston, MA, USA
| | | |
Collapse
|
5
|
Karal-ogly DD, Shumeev AN, Keburiya VV, Mintel MV, Rybtsov SA. Age-Related Changes in the Clustering of Blood Populations in Cynomolgus Monkeys Depend on Sex and Immune Status. Life (Basel) 2023; 13:life13020316. [PMID: 36836673 PMCID: PMC9965083 DOI: 10.3390/life13020316] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 01/24/2023] Open
Abstract
Non-anthropoid primates cynomolgus monkeys (Macaca fascicularis), also known as crab-eating macaques, are increasingly used in biomedical and preclinical studies due to their evolutionary proximity to humans, sharing similar diets, infectious and senile diseases. Age-related changes and sexual dimorphism of the immune system of C. monkeys have not been sufficiently characterized in literature, though age and sex differences affect the course of diseases and sensitivity to medications. Aging in C. monkeys is accompanied by an increase in CD3+CD4+CD8+ (DP-T) cells, plasma B-cells, and a decrease in platelets. Erythromyeloid bias has also been noticed in older animals. There was an increase in eosinophils, haematocrit (HCT) and haemoglobin concentration (HGB). Senile decline in the function of the immune system had sex differences. An increase in the number of monocytes, cytotoxic lymphocytes (CTL) and a decrease in the T-helper population were more pronounced in older females. A significant reduction in the number of B-cells and activated T-cells was detected in males only. A moderate correlation with the regression model of aging was established for DP-T, HCT and HGB. The reduction in the B cells count in males and the increase in CTL level in females are moderately correlated with age. Other blood cell populations did not show significant correlations in the regression models due to their high sample variability. The novel cell population CD3-CD20loCD16/CD56+, presumably NK-cells subset, was revealed. This cell population demonstrated an increase trend with age in both sexes. Population-statistical age norms for different sexes for young and very old macaques were established. The blood population clusters associated with sex and immune status in older animals were also identified.
Collapse
Affiliation(s)
| | - Alexander N. Shumeev
- Centre for Cell Technology and Immunology, Sirius University of Science and Technology, 354340 Sochi, Russia
| | | | - Marina V. Mintel
- The Research Institute of Medical Primatology, 354383 Sochi, Russia
| | - Stanislav A. Rybtsov
- Centre for Cell Technology and Immunology, Sirius University of Science and Technology, 354340 Sochi, Russia
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH8 9YL, UK
- Correspondence:
| |
Collapse
|
6
|
Wang MD, Tian J, Zhang JH, Zhao SY, Song MJ, Wang ZX. Human Galectin-7 Gene LGALS7 Promoter Sequence Polymorphisms and Risk of Spontaneous Intracerebral Hemorrhage: A Prospective Study. Front Mol Neurosci 2022; 15:840340. [PMID: 35401111 PMCID: PMC8984465 DOI: 10.3389/fnmol.2022.840340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 02/04/2022] [Indexed: 11/13/2022] Open
Abstract
Background and purposeDespite evidence for the role of genetic factors in stroke, only a small proportion of strokes have been clearly attributed to monogenic factors, due to phenotypic heterogeneity. The goal of this study was to determine whether a significant relationship exists between human galectin-7 gene LGALS7 promoter region polymorphisms and the risk of stroke due to non-traumatic intracerebral hemorrhage (ICH).MethodsThis two-stage genetic association study included an initial exploratory stage followed by a discovery stage. During the exploratory stage, transgenic galectin-7 mice or transgenic mice with the scrambled sequence of the hairpin structure –silenced down gene LGALS7—were generated and then expressed differentially expressed proteins and galectin-7-interacting proteins were identified through proteomic analysis. During the discovery stage, a single-nucleotide polymorphism (SNP) genotyping approach was used to determine associations between 2 LGALS7 SNPs and ICH stroke risk for a cohort of 24 patients with stroke of the Chinese Han population and 70 controls.ResultsDuring the exploratory phase, LGALS7 expression was found to be decreased in TGLGALS–DOWN mice as compared to its expression in TGLGALS mice. During the discovery phase, analysis of LGALS7 sequences of 24 non-traumatic ICH cases and 70 controls led to the identification of 2 ICH susceptibility loci: a genomic region on 19q13.2 containing two LGALS7 SNPs, rs567785577 and rs138945880, whereby the A allele of rs567785577 and the T allele of rs138945880 were associated with greater risk of contracting ICH [for T and A vs. C and G, unadjusted odds ratio (OR) = 13.5; 95% CI = 2.249–146.5; p = 0.002]. This is the first study to genotype the galectin-7 promoter in patients with hemorrhagic stroke. Genotype and allele association tests and preliminary analysis of patients with stroke revealed that a single locus may be a genetic risk factor for hemorrhagic stroke.ConclusionA and T alleles of two novel SNP loci of 19q13.2, rs567785577 and rs138945880, respectively, were evaluated for associations with susceptibility to ICH. Further studies with expanded case numbers that include subjects of other ethnic populations are needed to elucidate mechanisms underlying associations between these SNPs and ICH risk.
Collapse
Affiliation(s)
- Ming-Dong Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jing Tian
- Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, China National Clinical Research Center of Respiratory Disease, Beijing, China
| | - John H. Zhang
- Physiology Program, Department of Anesthesiology, Neurosurgery, Neurology, and Physiology, Center for Neuroscience Research, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Shun-Ying Zhao
- Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, China National Clinical Research Center of Respiratory Disease, Beijing, China
- *Correspondence: Shun-Ying Zhao,
| | - Ming-Jing Song
- Medical School, Huanghe Science and Technology University, Zhengzhou, China
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
- Ming-Jing Song,
| | - Zhan-Xiang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Zhan-Xiang Wang,
| |
Collapse
|
7
|
Izgi H, Han D, Isildak U, Huang S, Kocabiyik E, Khaitovich P, Somel M, Dönertaş HM. Inter-tissue convergence of gene expression during ageing suggests age-related loss of tissue and cellular identity. eLife 2022; 11:68048. [PMID: 35098922 PMCID: PMC8880995 DOI: 10.7554/elife.68048] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 01/28/2022] [Indexed: 11/13/2022] Open
Abstract
Developmental trajectories of gene expression may reverse in their direction during ageing, a phenomenon previously linked to cellular identity loss. Our analysis of cerebral cortex, lung, liver and muscle transcriptomes of 16 mice, covering development and ageing intervals, revealed widespread but tissue-specific ageing-associated expression reversals. Cumulatively, these reversals create a unique phenomenon: mammalian tissue transcriptomes diverge from each other during postnatal development, but during ageing, they tend to converge towards similar expression levels, a process we term Divergence followed by Convergence, or DiCo. We found that DiCo was most prevalent among tissue-specific genes and associated with loss of tissue identity, which is confirmed using data from independent mouse and human datasets. Further, using publicly available single-cell transcriptome data, we showed that DiCo could be driven both by alterations in tissue cell type composition and also by cell-autonomous expression changes within particular cell types.
Collapse
Affiliation(s)
- Hamit Izgi
- Department of Biological Sciences, Middle East Technical University, Ankara, Turkey
| | - DingDing Han
- CAS Key Laboratory of Computational Biology, Shanghai Institutes for Biological Sciences, Shanghai, China
| | - Ulas Isildak
- Department of Biological Sciences, Middle East Technical University, Ankara, Turkey
| | - Shuyun Huang
- CAS Key Laboratory of Computational Biology, Shanghai Institutes for Biological Sciences, Shanghai, China
| | - Ece Kocabiyik
- Department of Biological Sciences, Middle East Technical University, Ankara, Turkey
| | - Philipp Khaitovich
- Center for Neurobiology and Brain Restoration, Skolkovo Institute of Science and Technology, Moscow, Russian Federation
| | - Mehmet Somel
- Department of Biological Sciences, Middle East Technical University, Ankara, Turkey
| | | |
Collapse
|
8
|
Barth E, Srivastava A, Wengerodt D, Stojiljkovic M, Axer H, Witte OW, Kretz A, Marz M. Age-dependent expression changes of circadian system-related genes reveal a potentially conserved link to aging. Aging (Albany NY) 2021; 13:25694-25716. [PMID: 34923482 PMCID: PMC8751596 DOI: 10.18632/aging.203788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 12/01/2021] [Indexed: 12/12/2022]
Abstract
The circadian clock system influences the biology of life by establishing circadian rhythms in organisms, tissues, and cells, thus regulating essential biological processes based on the day/night cycle. Circadian rhythms change over a lifetime due to maturation and aging, and disturbances in the control of the circadian system are associated with several age-related pathologies. However, the impact of chronobiology and the circadian system on healthy organ and tissue aging remains largely unknown. Whether aging-related changes of the circadian system’s regulation follow a conserved pattern across different species and tissues, hence representing a common driving force of aging, is unclear. Based on a cross-sectional transcriptome analysis covering 329 RNA-Seq libraries, we provide indications that the circadian system is subjected to aging-related gene alterations shared between evolutionarily distinct species, such as Homo sapiens, Mus musculus, Danio rerio, and Nothobranchius furzeri. We discovered differentially expressed genes by comparing tissue-specific transcriptional profiles of mature, aged, and old-age individuals and report on six genes (per2, dec2, cirp, klf10, nfil3, and dbp) of the circadian system, which show conserved aging-related expression patterns in four organs of the species examined. Our results illustrate how the circadian system and aging might influence each other in various tissues over a long lifespan and conceptually complement previous studies tracking short-term diurnal and nocturnal gene expression oscillations.
Collapse
Affiliation(s)
- Emanuel Barth
- Bioinformatics/High Throughput Analysis, Faculty of Mathematics and Computer Science, Friedrich Schiller University Jena, Jena, Germany
| | - Akash Srivastava
- Bioinformatics/High Throughput Analysis, Faculty of Mathematics and Computer Science, Friedrich Schiller University Jena, Jena, Germany.,FLI Leibniz Institute for Age Research, Jena, Germany.,Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Diane Wengerodt
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Milan Stojiljkovic
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Hubertus Axer
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Otto W Witte
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Alexandra Kretz
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Manja Marz
- Bioinformatics/High Throughput Analysis, Faculty of Mathematics and Computer Science, Friedrich Schiller University Jena, Jena, Germany.,FLI Leibniz Institute for Age Research, Jena, Germany.,German Center for Integrative Biodiversity Research (iDiv), Halle-Jena-Leipzig, Germany.,European Virus Bioinformatics Center (EVBC), Jena, Germany
| |
Collapse
|
9
|
Ferreira M, Francisco S, Soares AR, Nobre A, Pinheiro M, Reis A, Neto S, Rodrigues AJ, Sousa N, Moura G, Santos MAS. Integration of segmented regression analysis with weighted gene correlation network analysis identifies genes whose expression is remodeled throughout physiological aging in mouse tissues. Aging (Albany NY) 2021; 13:18150-18190. [PMID: 34330884 PMCID: PMC8351669 DOI: 10.18632/aging.203379] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 07/21/2021] [Indexed: 02/06/2023]
Abstract
Gene expression alterations occurring with aging have been described for a multitude of species, organs, and cell types. However, most of the underlying studies rely on static comparisons of mean gene expression levels between age groups and do not account for the dynamics of gene expression throughout the lifespan. These studies also tend to disregard the pairwise relationships between gene expression profiles, which may underlie commonly altered pathways and regulatory mechanisms with age. To overcome these limitations, we have combined segmented regression analysis with weighted gene correlation network analysis (WGCNA) to identify high-confidence signatures of aging in the brain, heart, liver, skeletal muscle, and pancreas of C57BL/6 mice in a publicly available RNA-Seq dataset (GSE132040). Functional enrichment analysis of the overlap of genes identified in both approaches showed that immune- and inflammation-related responses are prominently altered in the brain and the liver, while in the heart and the muscle, aging affects amino and fatty acid metabolism, and tissue regeneration, respectively, which reflects an age-related global loss of tissue function. We also explored sexual dimorphism in the aging mouse transcriptome and found the liver and the muscle to have the most pronounced gender differences in gene expression throughout the lifespan, particularly in proteostasis-related pathways. While the data showed little overlap among the age-dysregulated genes between tissues, aging triggered common biological processes in distinct tissues, which we highlight as important features of murine tissue physiological aging.
Collapse
Affiliation(s)
- Margarida Ferreira
- Institute of Biomedicine – iBiMED, Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
| | - Stephany Francisco
- Institute of Biomedicine – iBiMED, Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
| | - Ana R. Soares
- Institute of Biomedicine – iBiMED, Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
| | - Ana Nobre
- Institute of Biomedicine – iBiMED, Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
| | - Miguel Pinheiro
- Institute of Biomedicine – iBiMED, Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
| | - Andreia Reis
- Institute of Biomedicine – iBiMED, Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
| | - Sonya Neto
- Institute of Biomedicine – iBiMED, Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
| | - Ana João Rodrigues
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga 4710-057, Portugal
- ICVS/3B’s–PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga 4710-057, Portugal
- ICVS/3B’s–PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Gabriela Moura
- Institute of Biomedicine – iBiMED, Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
| | - Manuel A. S. Santos
- Institute of Biomedicine – iBiMED, Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
| |
Collapse
|
10
|
Takemon Y, Chick JM, Gerdes Gyuricza I, Skelly DA, Devuyst O, Gygi SP, Churchill GA, Korstanje R. Proteomic and transcriptomic profiling reveal different aspects of aging in the kidney. eLife 2021; 10:e62585. [PMID: 33687326 PMCID: PMC8096428 DOI: 10.7554/elife.62585] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 03/06/2021] [Indexed: 01/10/2023] Open
Abstract
Little is known about the molecular changes that take place in the kidney during the aging process. In order to better understand these changes, we measured mRNA and protein levels in genetically diverse mice at different ages. We observed distinctive change in mRNA and protein levels as a function of age. Changes in both mRNA and protein are associated with increased immune infiltration and decreases in mitochondrial function. Proteins show a greater extent of change and reveal changes in a wide array of biological processes including unique, organ-specific features of aging in kidney. Most importantly, we observed functionally important age-related changes in protein that occur in the absence of corresponding changes in mRNA. Our findings suggest that mRNA profiling alone provides an incomplete picture of molecular aging in the kidney and that examination of changes in proteins is essential to understand aging processes that are not transcriptionally regulated.
Collapse
Affiliation(s)
| | - Joel M Chick
- Harvard Medical SchoolBostonUnited States
- VividionTherapeuticsSan DiegoUnited States
| | | | | | - Olivier Devuyst
- Institute of Physiology, University of ZurichZurichSwitzerland
| | | | | | | |
Collapse
|
11
|
Fuentealba M, Fabian DK, Dönertaş HM, Thornton JM, Partridge L. Transcriptomic profiling of long- and short-lived mutant mice implicates mitochondrial metabolism in ageing and shows signatures of normal ageing in progeroid mice. Mech Ageing Dev 2021; 194:111437. [PMID: 33454277 PMCID: PMC7895802 DOI: 10.1016/j.mad.2021.111437] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 12/09/2020] [Accepted: 01/11/2021] [Indexed: 12/21/2022]
Abstract
Genetically modified mouse models of ageing are the living proof that lifespan and healthspan can be lengthened or shortened, and provide a powerful context in which to unravel the molecular mechanisms at work. In this study, we analysed and compared gene expression data from 10 long-lived and 8 short-lived mouse models of ageing. Transcriptome-wide correlation analysis revealed that mutations with equivalent effects on lifespan induce more similar transcriptomic changes, especially if they target the same pathway. Using functional enrichment analysis, we identified 58 gene sets with consistent changes in long- and short-lived mice, 55 of which were up-regulated in long-lived mice and down-regulated in short-lived mice. Half of these sets represented genes involved in energy and lipid metabolism, among which Ppargc1a, Mif, Aldh5a1 and Idh1 were frequently observed. Based on the gene sets with consistent changes, and also the whole transcriptome, the gene expression changes during normal ageing resembled the transcriptome of short-lived models, suggesting that accelerated ageing models reproduce partially the molecular changes of ageing. Finally, we identified new genetic interventions that may ameliorate ageing, by comparing the transcriptomes of 51 mouse mutants not previously associated with ageing to expression signatures of long- and short-lived mice and ageing-related changes.
Collapse
Affiliation(s)
- Matias Fuentealba
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, London, WC1E 6BT, UK; European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, CB10 1SD, UK
| | - Daniel K Fabian
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, London, WC1E 6BT, UK; European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, CB10 1SD, UK
| | - Handan Melike Dönertaş
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, CB10 1SD, UK
| | - Janet M Thornton
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, CB10 1SD, UK
| | - Linda Partridge
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, London, WC1E 6BT, UK; Max Planck Institute for Biology of Ageing, Cologne, Germany.
| |
Collapse
|
12
|
Shafiq K, Sanghai N, Guo Y, Kong J. Implication of post-translationally modified SOD1 in pathological aging. GeroScience 2021; 43:507-515. [PMID: 33608813 PMCID: PMC8110659 DOI: 10.1007/s11357-021-00332-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 01/31/2021] [Indexed: 12/18/2022] Open
Abstract
Why certain people relish healthy aging throughout their life span while others suffer pathological consequences? In this review, we focus on some of the dominant paradigms of pathological aging, such as amyotrophic lateral sclerosis (ALS), Alzheimer's disease (AD), and Parkinson's disease (PD), and predict that the antioxidant superoxide dismutase 1 (SOD1), when post-translationally modified by aging-associated oxidative stress, acts as a mechanism to accelerated aging in these age-related neurodegenerative diseases. Oxidative modifications of natively reduced SOD1 induce pathological confirmations such as misfolding, leading to a subsequent formation of monomeric, oligomeric, and multimeric aggregates. Misfolded SOD1 propagates like prions from cell to cell. These modified conformations are detected in brain tissues in ALS, AD, and PD, and are considered a contributing factor to their initial pathogenesis. We have also elaborated on oxidative stress-induced non-native modifications of SOD1 and offered a logistic argument on their global implication in accelerated or pathological aging in the context of ALS, AD, and PD.
Collapse
Affiliation(s)
- Kashfia Shafiq
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Science, University of Manitoba, Winnipeg, MB, R3E 0J9, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, 27, King's College Cir, Toronto, ON, M5S, Canada
| | - Nitesh Sanghai
- College of Pharmacy, Rady Faculty of Health Science, University of Manitoba, Winnipeg, MB, R3E 0T5, Canada
| | - Ying Guo
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Science, University of Manitoba, Winnipeg, MB, R3E 0J9, Canada.,Pathological Department, Hebei North University, Zhangjiakou, Hebei, China
| | - Jiming Kong
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Science, University of Manitoba, Winnipeg, MB, R3E 0J9, Canada.
| |
Collapse
|
13
|
Tissue-specific Gene Expression Changes Are Associated with Aging in Mice. GENOMICS PROTEOMICS & BIOINFORMATICS 2020; 18:430-442. [PMID: 33309863 PMCID: PMC8242333 DOI: 10.1016/j.gpb.2020.12.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 03/13/2019] [Accepted: 04/10/2019] [Indexed: 12/17/2022]
Abstract
Aging is a complex process that can be characterized by functional and cognitive decline in an individual. Aging can be assessed based on the functional capacity of vital organs and their intricate interactions with one another. Thus, the nature of aging can be described by focusing on a specific organ and an individual itself. However, to fully understand the complexity of aging, one must investigate not only a single tissue or biological process but also its complex interplay and interdependencies with other biological processes. Here, using RNA-seq, we monitored changes in the transcriptome during aging in four tissues (including brain, blood, skin and liver) in mice at 9 months, 15 months, and 24 months, with a final evaluation at the very old age of 30 months. We identified several genes and processes that were differentially regulated during aging in both tissue-dependent and tissue-independent manners. Most importantly, we found that the electron transport chain (ETC) of mitochondria was similarly affected at the transcriptome level in the four tissues during the aging process. We also identified the liver as the tissue showing the largest variety of differentially expressed genes (DEGs) over time. Lcn2 (Lipocalin-2) was found to be similarly regulated among all tissues, and its effect on longevity and survival was validated using its orthologue in Caenorhabditis elegans. Our study demonstrated that the molecular processes of aging are relatively subtle in their progress, and the aging process of every tissue depends on the tissue’s specialized function and environment. Hence, individual gene or process alone cannot be described as the key of aging in the whole organism.
Collapse
|
14
|
Smith JC, Sausville EL, Girish V, Yuan ML, Vasudevan A, John KM, Sheltzer JM. Cigarette Smoke Exposure and Inflammatory Signaling Increase the Expression of the SARS-CoV-2 Receptor ACE2 in the Respiratory Tract. Dev Cell 2020; 53:514-529.e3. [PMID: 32425701 PMCID: PMC7229915 DOI: 10.1016/j.devcel.2020.05.012] [Citation(s) in RCA: 280] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/26/2020] [Accepted: 05/12/2020] [Indexed: 01/06/2023]
Abstract
The factors mediating fatal SARS-CoV-2 infections are poorly understood. Here, we show that cigarette smoke causes a dose-dependent upregulation of angiotensin converting enzyme 2 (ACE2), the SARS-CoV-2 receptor, in rodent and human lungs. Using single-cell sequencing data, we demonstrate that ACE2 is expressed in a subset of secretory cells in the respiratory tract. Chronic smoke exposure triggers the expansion of this cell population and a concomitant increase in ACE2 expression. In contrast, quitting smoking decreases the abundance of these secretory cells and reduces ACE2 levels. Finally, we demonstrate that ACE2 expression is responsive to inflammatory signaling and can be upregulated by viral infections or interferon treatment. Taken together, these results may partially explain why smokers are particularly susceptible to severe SARS-CoV-2 infections. Furthermore, our work identifies ACE2 as an interferon-stimulated gene in lung cells, suggesting that SARS-CoV-2 infections could create positive feedback loops that increase ACE2 levels and facilitate viral dissemination.
Collapse
Affiliation(s)
- Joan C Smith
- Google, Inc., New York City, NY 10011, USA; Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Erin L Sausville
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Vishruth Girish
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; Stony Brook University, Stony Brook, NY 11794, USA
| | - Monet Lou Yuan
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; Johns Hopkins University, Baltimore, MD 21218, USA
| | - Anand Vasudevan
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Kristen M John
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; Hofstra University, Hempstead, NY 11549, USA
| | - Jason M Sheltzer
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
15
|
Rowland J, Akbarov A, Eales J, Xu X, Dormer JP, Guo H, Denniff M, Jiang X, Ranjzad P, Nazgiewicz A, Prestes PR, Antczak A, Szulinska M, Wise IA, Zukowska-Szczechowska E, Bogdanski P, Woolf AS, Samani NJ, Charchar FJ, Tomaszewski M. Uncovering genetic mechanisms of kidney aging through transcriptomics, genomics, and epigenomics. Kidney Int 2020; 95:624-635. [PMID: 30784661 PMCID: PMC6390171 DOI: 10.1016/j.kint.2018.10.029] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 10/15/2018] [Accepted: 10/18/2018] [Indexed: 12/19/2022]
Abstract
Nephrons scar and involute during aging, increasing the risk of chronic kidney disease. Little is known, however, about genetic mechanisms of kidney aging. We sought to define the signatures of age on the renal transcriptome using 563 human kidneys. The initial discovery analysis of 260 kidney transcriptomes from the TRANScriptome of renaL humAn TissuE Study (TRANSLATE) and the Cancer Genome Atlas identified 37 age-associated genes. For 19 of those genes, the association with age was replicated in 303 kidney transcriptomes from the Nephroseq resource. Surveying 42 nonrenal tissues from the Genotype–Tissue Expression project revealed that, for approximately a fifth of the replicated genes, the association with age was kidney-specific. Seventy-three percent of the replicated genes were associated with functional or histological parameters of age-related decline in kidney health, including glomerular filtration rate, glomerulosclerosis, interstitial fibrosis, tubular atrophy, and arterial narrowing. Common genetic variants in four of the age-related genes, namely LYG1, PPP1R3C, LTF and TSPYL5, correlated with the trajectory of age-related changes in their renal expression. Integrative analysis of genomic, epigenomic, and transcriptomic information revealed that the observed age-related decline in renal TSPYL5 expression was determined both genetically and epigenetically. Thus, this study revealed robust molecular signatures of the aging kidney and new regulatory mechanisms of age-related change in the kidney transcriptome.
Collapse
Affiliation(s)
- Joshua Rowland
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Artur Akbarov
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - James Eales
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Xiaoguang Xu
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - John P Dormer
- Department of Cellular Pathology, University Hospitals of Leicester, Leicester, UK
| | - Hui Guo
- Division of Population Health, Health Services Research and Primary Care, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Matthew Denniff
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Xiao Jiang
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Parisa Ranjzad
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
| | - Alicja Nazgiewicz
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | | | - Andrzej Antczak
- Department of Urology and Uro-oncology, Karol Marcinkowski University of Medical Sciences, Poznan, Poland
| | - Monika Szulinska
- Department of Treatment of Obesity, Metabolic Disorders and Clinical Dietetics, Poznan University of Medical Sciences, Poznan, Poland
| | - Ingrid A Wise
- Faculty of Health and Life Sciences, Federation University Australia, Ballarat, Victoria, Australia
| | - Ewa Zukowska-Szczechowska
- Department of Health Care, Silesian Medical College, Katowice, Poland; Department of Internal Medicine, Diabetology and Nephrology, Medical University of Silesia, Zabrze, Poland
| | - Pawel Bogdanski
- Department of Treatment of Obesity, Metabolic Disorders and Clinical Dietetics, Poznan University of Medical Sciences, Poznan, Poland
| | - Adrian S Woolf
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK; Department of Paediatric Nephrology, Royal Manchester Children's Hospital, Manchester University National Health Service Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK; Leicester National Institute for Health Research Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Fadi J Charchar
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK; Faculty of Health and Life Sciences, Federation University Australia, Ballarat, Victoria, Australia; Department of Physiology, University of Melbourne, Parkville, Victoria, Australia
| | - Maciej Tomaszewski
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK; Division of Medicine and Manchester Heart Centre, Manchester University National Health Service Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK.
| |
Collapse
|
16
|
Kimmel JC, Penland L, Rubinstein ND, Hendrickson DG, Kelley DR, Rosenthal AZ. Murine single-cell RNA-seq reveals cell-identity- and tissue-specific trajectories of aging. Genome Res 2019; 29:2088-2103. [PMID: 31754020 PMCID: PMC6886498 DOI: 10.1101/gr.253880.119] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 10/21/2019] [Indexed: 01/08/2023]
Abstract
Aging is a pleiotropic process affecting many aspects of mammalian physiology. Mammals are composed of distinct cell type identities and tissue environments, but the influence of these cell identities and environments on the trajectory of aging in individual cells remains unclear. Here, we performed single-cell RNA-seq on >50,000 individual cells across three tissues in young and old mice to allow for direct comparison of aging phenotypes across cell types. We found transcriptional features of aging common across many cell types, as well as features of aging unique to each type. Leveraging matrix factorization and optimal transport methods, we found that both cell identities and tissue environments exert influence on the trajectory and magnitude of aging, with cell identity influence predominating. These results suggest that aging manifests with unique directionality and magnitude across the diverse cell identities in mammals.
Collapse
Affiliation(s)
- Jacob C Kimmel
- Calico Life Sciences, South San Francisco, California 94080, USA
| | - Lolita Penland
- Calico Life Sciences, South San Francisco, California 94080, USA
| | | | | | - David R Kelley
- Calico Life Sciences, South San Francisco, California 94080, USA
| | - Adam Z Rosenthal
- Calico Life Sciences, South San Francisco, California 94080, USA
| |
Collapse
|
17
|
Usui Y, Kimura Y, Satoh T, Takemura N, Ouchi Y, Ohmiya H, Kobayashi K, Suzuki H, Koyama S, Hagiwara S, Tanaka H, Imoto S, Eberl G, Asami Y, Fujimoto K, Uematsu S. Effects of long-term intake of a yogurt fermented with Lactobacillus delbrueckii subsp. bulgaricus 2038 and Streptococcus thermophilus 1131 on mice. Int Immunol 2019; 30:319-331. [PMID: 29767727 DOI: 10.1093/intimm/dxy035] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 05/12/2018] [Indexed: 01/12/2023] Open
Abstract
The gut is an extremely complicated ecosystem where micro-organisms, nutrients and host cells interact vigorously. Although the function of the intestine and its barrier system weakens with age, some probiotics can potentially prevent age-related intestinal dysfunction. Lactobacillus delbrueckii subsp. bulgaricus 2038 and Streptococcus thermophilus 1131, which are the constituents of LB81 yogurt, are representative probiotics. However, it is unclear whether their long-term intake has a beneficial influence on systemic function. Here, we examined the gut microbiome, fecal metabolites and gene expression profiles of various organs in mice. Although age-related alterations were apparent in them, long-term LB81 yogurt intake led to an increased Bacteroidetes to Firmicutes ratio and elevated abundance of the bacterial family S24-7 (Bacteroidetes), which is known to be associated with butyrate and propanoate production. According to our fecal metabolite analysis to detect enrichment, long-term LB81 yogurt intake altered the intestinal metabolic pathways associated with propanoate and butanoate in the mice. Gene ontology analysis also revealed that long-term LB81 yogurt intake influenced many physiological functions related to the defense response. The profiles of various genes associated with antimicrobial peptides-, tight junctions-, adherens junctions- and mucus-associated intestinal barrier functions were also drastically altered in the LB81 yogurt-fed mice. Thus, long-term intake of LB81 yogurt has the potential to maintain systemic homeostasis, such as the gut barrier function, by controlling the intestinal microbiome and its metabolites.
Collapse
Affiliation(s)
- Yuki Usui
- Division of Systems Immunology, The Institute of Medical Science, University of Tokyo, Shirokanedai, Minato-ku, Tokyo, Japan
| | - Yasumasa Kimura
- Division of Systems Immunology, The Institute of Medical Science, University of Tokyo, Shirokanedai, Minato-ku, Tokyo, Japan
| | - Takeshi Satoh
- Division of Systems Immunology, The Institute of Medical Science, University of Tokyo, Shirokanedai, Minato-ku, Tokyo, Japan
| | - Naoki Takemura
- Department of Mucosal Immunology, School of Medicine, Chiba University, Inohana, Chuou-ku, Chiba, Japan.,Division of Innate Immune Regulation, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, University of Tokyo, Shirokanedai, Minato-ku, Tokyo, Japan
| | - Yasuo Ouchi
- Department of Mucosal Immunology, School of Medicine, Chiba University, Inohana, Chuou-ku, Chiba, Japan
| | - Hiroko Ohmiya
- Division of Systems Immunology, The Institute of Medical Science, University of Tokyo, Shirokanedai, Minato-ku, Tokyo, Japan
| | - Kyosuke Kobayashi
- Food Science Research Laboratories, R&D Division, Meiji Co., Ltd, Naruda, Odawara, Kanagawa, Japan
| | - Hiromi Suzuki
- Food Science Research Laboratories, R&D Division, Meiji Co., Ltd, Naruda, Odawara, Kanagawa, Japan
| | - Satomi Koyama
- Food Science Research Laboratories, R&D Division, Meiji Co., Ltd, Naruda, Odawara, Kanagawa, Japan
| | - Satoko Hagiwara
- Food Science Research Laboratories, R&D Division, Meiji Co., Ltd, Naruda, Odawara, Kanagawa, Japan
| | - Hirotoshi Tanaka
- Division of Rheumatology, Center for Antibody and Vaccine Therapy, IMSUT Hospital, The Institute of Medical Science, University of Tokyo, Shirokanedai, Minato-ku, Tokyo, Japan
| | - Seiya Imoto
- Division of Health Medical Data Science, Health Intelligence Center, The Institute of Medical Science, University of Tokyo, Shirokanedai, Minato-ku, Tokyo, Japan
| | - Gérard Eberl
- Institut Pasteur, Microenvironment and Immunity Unit, Paris, France.,INSERM, Paris, France
| | - Yukio Asami
- Food Science Research Laboratories, R&D Division, Meiji Co., Ltd, Naruda, Odawara, Kanagawa, Japan
| | - Kosuke Fujimoto
- Department of Mucosal Immunology, School of Medicine, Chiba University, Inohana, Chuou-ku, Chiba, Japan.,Division of Innate Immune Regulation, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, University of Tokyo, Shirokanedai, Minato-ku, Tokyo, Japan
| | - Satoshi Uematsu
- Department of Mucosal Immunology, School of Medicine, Chiba University, Inohana, Chuou-ku, Chiba, Japan.,Division of Innate Immune Regulation, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, University of Tokyo, Shirokanedai, Minato-ku, Tokyo, Japan
| |
Collapse
|
18
|
Turan ZG, Parvizi P, Dönertaş HM, Tung J, Khaitovich P, Somel M. Molecular footprint of Medawar's mutation accumulation process in mammalian aging. Aging Cell 2019; 18:e12965. [PMID: 31062469 PMCID: PMC6612638 DOI: 10.1111/acel.12965] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 02/14/2019] [Accepted: 03/28/2019] [Indexed: 12/20/2022] Open
Abstract
Medawar's mutation accumulation hypothesis explains aging by the declining force of natural selection with age: Slightly deleterious germline mutations expressed in old age can drift to fixation and thereby lead to aging‐related phenotypes. Although widely cited, empirical evidence for this hypothesis has remained limited. Here, we test one of its predictions that genes relatively highly expressed in old adults should be under weaker purifying selection than genes relatively highly expressed in young adults. Combining 66 transcriptome datasets (including 16 tissues from five mammalian species) with sequence conservation estimates across mammals, here we report that the overall conservation level of expressed genes is lower at old age compared to young adulthood. This age‐related decrease in transcriptome conservation (ADICT) is systematically observed in diverse mammalian tissues, including the brain, liver, lung, and artery, but not in others, most notably in the muscle and heart. Where observed, ADICT is driven partly by poorly conserved genes being up‐regulated during aging. In general, the more often a gene is found up‐regulated with age among tissues and species, the lower its evolutionary conservation. Poorly conserved and up‐regulated genes have overlapping functional properties that include responses to age‐associated tissue damage, such as apoptosis and inflammation. Meanwhile, these genes do not appear to be under positive selection. Hence, genes contributing to old age phenotypes are found to harbor an excess of slightly deleterious alleles, at least in certain tissues. This supports the notion that genetic drift shapes aging in multicellular organisms, consistent with Medawar's mutation accumulation hypothesis.
Collapse
Affiliation(s)
- Zeliha Gözde Turan
- Department of Biological Sciences Middle East Technical University Ankara Turkey
| | - Poorya Parvizi
- Department of Biological Sciences Middle East Technical University Ankara Turkey
- Usher Institute of Population Health Sciences and Informatics University of Edinburgh Edinburgh UK
| | - Handan Melike Dönertaş
- European Molecular Biology Laboratory, European Bioinformatics Institute EMBL‐EBI Wellcome Trust Genome Campus Cambridge UK
| | - Jenny Tung
- Department of Evolutionary Anthropology Duke University Durham North Carolina
- Department of Biology Duke University Durham North Carolina
- Duke Population Research Institute Duke University Durham North Carolina
| | - Philipp Khaitovich
- Center for Neurobiology and Brain Restoration Skolkovo Institute of Science and Technology Moscow Russia
- CAS Key Laboratory of Computational Biology, CAS‐MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences Chinese Academy of Sciences Shanghai China
| | - Mehmet Somel
- Department of Biological Sciences Middle East Technical University Ankara Turkey
| |
Collapse
|
19
|
Ortega-Martínez M, Gutiérrez-Dávila V, Niderhauser-García A, Cerda-Flores RM, García-Juárez J, de-la-Garza-González C, Jaramillo-Rangel G. Morphometric analysis of the non-epithelial areas of mouse bronchioles through the normal aging process. Am J Transl Res 2019; 11:3637-3644. [PMID: 31312374 PMCID: PMC6614624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 05/25/2019] [Indexed: 06/10/2023]
Abstract
Aging is associated with changes in the structure and function of the lung that may increase susceptibility to chronic lung diseases. The aim of this study was the morphometric assessment of the non-epithelial areas of the bronchioles of mouse through the normal aging process. Lungs from CD1 mice at the age of 2, 6, 12, 18, or 24 months were fixed in neutral-buffered formalin and paraffin-embedded. Sections were cut, stained with Masson trichrome, and examined using a light microscope. High-resolution color images were captured using a camera linked to image analysis software to measure areas and lengths. We observed in the bronchioles through the aging process an increase of the total area, an increase of the lumen area, and a decrease of the wall area. In conclusion, our results revealed structural changes in the bronchioles of mouse through the normal aging process. These alterations are likely to contribute to development of chronic lung diseases.
Collapse
Affiliation(s)
- Marta Ortega-Martínez
- Department of Pathology, School of Medicine, Autonomous University of Nuevo LeonMonterrey 64460, Mexico
| | - Vanessa Gutiérrez-Dávila
- Department of Pathology, School of Medicine, Autonomous University of Nuevo LeonMonterrey 64460, Mexico
| | | | | | - Jaime García-Juárez
- Department of Histology, School of Medicine, Autonomous University of Nuevo LeonMonterrey 64460, Mexico
| | | | - Gilberto Jaramillo-Rangel
- Department of Pathology, School of Medicine, Autonomous University of Nuevo LeonMonterrey 64460, Mexico
| |
Collapse
|
20
|
Yoon SK, Okyere BA, Strasser D. Polypharmacy and Rational Prescribing: Changing the Culture of Medicine One Patient at a Time. CURRENT PHYSICAL MEDICINE AND REHABILITATION REPORTS 2019. [DOI: 10.1007/s40141-019-00220-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
21
|
Partearroyo T, Murillo-Cuesta S, Vallecillo N, Bermúdez-Muñoz JM, Rodríguez-de la Rosa L, Mandruzzato G, Celaya AM, Zeisel SH, Pajares MA, Varela-Moreiras G, Varela-Nieto I. Betaine-homocysteine S-methyltransferase deficiency causes increased susceptibility to noise-induced hearing loss associated with plasma hyperhomocysteinemia. FASEB J 2019; 33:5942-5956. [PMID: 30753104 PMCID: PMC6463923 DOI: 10.1096/fj.201801533r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 01/15/2019] [Indexed: 12/16/2022]
Abstract
Betaine-homocysteine S-methyltransferases (BHMTs) are methionine cycle enzymes that remethylate homocysteine; hence, their malfunction leads to hyperhomocysteinemia. Epidemiologic and experimental studies have revealed a correlation between hyperhomocysteinemia and hearing loss. Here, we have studied the expression of methionine cycle genes in the mouse cochlea and the impact of knocking out the Bhmt gene in the auditory receptor. We evaluated age-related changes in mouse hearing by recording auditory brainstem responses before and following exposure to noise. Also, we measured cochlear cytoarchitecture, gene expression by RNA-arrays and quantitative RT-PCR, and metabolite levels in liver and plasma by HPLC. Our results indicate that there is an age-dependent strain-specific expression of methionine cycle genes in the mouse cochlea and a further regulation during the response to noise damage. Loss of Bhmt did not cause an evident impact in the hearing acuity of young mice, but it produced higher threshold shifts and poorer recovery following noise challenge. Hearing loss was associated with increased cochlear injury, outer hair cell loss, altered expression of cochlear methionine cycle genes, and hyperhomocysteinemia. Our results suggest that BHMT plays a central role in the homeostasis of cochlear methionine metabolism and that Bhmt2 up-regulation could carry out a compensatory role in cochlear protection against noise injury in the absence of BHMT.-Partearroyo, T., Murillo-Cuesta, S., Vallecillo, N., Bermúdez-Muñoz, J. M., Rodríguez-de la Rosa, L., Mandruzzato, G., Celaya, A. M., Zeisel, S. H., Pajares, M. A., Varela-Moreiras, G., Varela-Nieto, I. Betaine-homocysteine S-methyltransferase deficiency causes increased susceptibility to noise-induced hearing loss associated with plasma hyperhomocysteinemia.
Collapse
Affiliation(s)
- Teresa Partearroyo
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad Centro de Estudios Universitarios CEU San Pablo, Madrid, Spain
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas–Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
| | - Silvia Murillo-Cuesta
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas–Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigación Sanitaria La Paz (IdiPAZ), Madrid, Spain
| | - Néstor Vallecillo
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas–Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
| | - Jose M. Bermúdez-Muñoz
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas–Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Lourdes Rodríguez-de la Rosa
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas–Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigación Sanitaria La Paz (IdiPAZ), Madrid, Spain
| | | | - Adelaida M. Celaya
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas–Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Steven H. Zeisel
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina, USA; and
| | - María A. Pajares
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas–Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
- Instituto de Investigación Sanitaria La Paz (IdiPAZ), Madrid, Spain
- Centro de Investigaciones Biológicas, (CSIC) Madrid, Spain
| | - Gregorio Varela-Moreiras
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad Centro de Estudios Universitarios CEU San Pablo, Madrid, Spain
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas–Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
| | - Isabel Varela-Nieto
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas–Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigación Sanitaria La Paz (IdiPAZ), Madrid, Spain
| |
Collapse
|
22
|
Fernando R, Drescher C, Deubel S, Jung T, Ost M, Klaus S, Grune T, Castro JP. Low proteasomal activity in fast skeletal muscle fibers is not associated with increased age-related oxidative damage. Exp Gerontol 2019; 117:45-52. [DOI: 10.1016/j.exger.2018.10.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 10/09/2018] [Accepted: 10/23/2018] [Indexed: 01/07/2023]
|
23
|
Fernando R, Drescher C, Nowotny K, Grune T, Castro JP. Impaired proteostasis during skeletal muscle aging. Free Radic Biol Med 2019; 132:58-66. [PMID: 30194981 DOI: 10.1016/j.freeradbiomed.2018.08.037] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 08/03/2018] [Accepted: 08/30/2018] [Indexed: 01/03/2023]
Abstract
Aging is a complex phenomenon that has detrimental effects on tissue homeostasis. The skeletal muscle is one of the earliest tissues to be affected and to manifest age-related changes such as functional impairment and the loss of mass. Common to these alterations and to most of tissues during aging is the disruption of the proteostasis network by detrimental changes in the ubiquitin-proteasomal system (UPS) and the autophagy-lysosomal system (ALS). In fact, during aging the accumulation of protein aggregates, a process mainly driven by increased levels of oxidative stress, has been observed, clearly demonstrating UPS and ALS dysregulation. Since the UPS and ALS are the two most important pathways for the removal of misfolded and aggregated proteins and also of damaged organelles, we provide here an overview on the current knowledge regarding the connection between the loss of proteostasis and skeletal muscle functional impairment and also how redox regulation can play a role during aging. Therefore, this review serves for a better understanding of skeletal muscle aging in regard to the loss of proteostasis and how redox regulation can impact its function and maintenance.
Collapse
Affiliation(s)
- Raquel Fernando
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbrücke, 14558 Nuthetal, Germany
| | - Cathleen Drescher
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbrücke, 14558 Nuthetal, Germany
| | - Kerstin Nowotny
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbrücke, 14558 Nuthetal, Germany
| | - Tilman Grune
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbrücke, 14558 Nuthetal, Germany; German Center for Diabetes Research (DZD), 85764 München-Neuherberg, Germany; German Center for Cardiovascular Research (DZHK), 10117 Berlin, Germany; University of Potsdam, Institute of Nutritional Science, 14558 Nuthetal, Germany
| | - José Pedro Castro
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbrücke, 14558 Nuthetal, Germany; German Center for Diabetes Research (DZD), 85764 München-Neuherberg, Germany; Faculty of Medicine, Department for Biomedicine, University of Porto, 4200-319, Portugal; Institute for Innovation and Health Research (I3S), Aging and Stress Group, R. Alfredo Allen, 4200-135 Porto, Portugal.
| |
Collapse
|
24
|
Voic H, Li X, Jang JH, Zou C, Sundd P, Alder J, Rojas M, Chandra D, Randell S, Mallampalli RK, Tesfaigzi Y, Ryba T, Nyunoya T. RNA sequencing identifies common pathways between cigarette smoke exposure and replicative senescence in human airway epithelia. BMC Genomics 2019; 20:22. [PMID: 30626320 PMCID: PMC6325884 DOI: 10.1186/s12864-018-5409-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 12/26/2018] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Aging is affected by genetic and environmental factors, and cigarette smoking is strongly associated with accumulation of senescent cells. In this study, we wanted to identify genes that may potentially be beneficial for cell survival in response to cigarette smoke and thereby may contribute to development of cellular senescence. RESULTS Primary human bronchial epithelial cells from five healthy donors were cultured, treated with or without 1.5% cigarette smoke extract (CSE) for 24 h or were passaged into replicative senescence. Transcriptome changes were monitored using RNA-seq in CSE and non-CSE exposed cells and those passaged into replicative senescence. We found that, among 1534 genes differentially regulated during senescence and 599 after CSE exposure, 243 were altered in both conditions, representing strong enrichment. Pathways and gene sets overrepresented in both conditions belonged to cellular processes that regulate reactive oxygen species, proteasome degradation, and NF-κB signaling. CONCLUSIONS Our results offer insights into gene expression responses during cellular aging and cigarette smoke exposure, and identify potential molecular pathways that are altered by cigarette smoke and may also promote airway epithelial cell senescence.
Collapse
Affiliation(s)
- Hannah Voic
- 0000 0004 0504 9575grid.422569.eDivision of Natural Sciences, New College of Florida, Sarasota, FL USA
| | - Xiuying Li
- 0000 0004 1936 9000grid.21925.3dDepartment of Medicine, University of Pittsburgh, NW628 UPMC Montefiore, 3459 Fifth Avenue, Pittsburgh, PA 15213 USA ,0000 0004 0420 3665grid.413935.9VA Pittsburgh Healthcare System, Pittsburgh, PA USA
| | - Jun-Ho Jang
- 0000 0004 0454 5075grid.417046.0Cardiovascular Institute, Department of Medicine, Allegheny Health Network, Pittsburgh, PA USA
| | - Chunbin Zou
- 0000 0004 1936 9000grid.21925.3dDepartment of Medicine, University of Pittsburgh, NW628 UPMC Montefiore, 3459 Fifth Avenue, Pittsburgh, PA 15213 USA ,0000 0004 0420 3665grid.413935.9VA Pittsburgh Healthcare System, Pittsburgh, PA USA
| | - Prithu Sundd
- 0000 0004 1936 9000grid.21925.3dVascular Medicine Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, PA USA
| | - Jonathan Alder
- 0000 0004 1936 9000grid.21925.3dDepartment of Medicine, University of Pittsburgh, NW628 UPMC Montefiore, 3459 Fifth Avenue, Pittsburgh, PA 15213 USA
| | - Mauricio Rojas
- 0000 0004 1936 9000grid.21925.3dDepartment of Medicine, University of Pittsburgh, NW628 UPMC Montefiore, 3459 Fifth Avenue, Pittsburgh, PA 15213 USA
| | - Divay Chandra
- 0000 0004 1936 9000grid.21925.3dDepartment of Medicine, University of Pittsburgh, NW628 UPMC Montefiore, 3459 Fifth Avenue, Pittsburgh, PA 15213 USA
| | - Scott Randell
- 0000 0001 1034 1720grid.410711.2Department of Cell and Molecular Physiology, University of North Carolina, Chapel Hill, NC USA
| | - Rama K. Mallampalli
- 0000 0004 1936 9000grid.21925.3dDepartment of Medicine, University of Pittsburgh, NW628 UPMC Montefiore, 3459 Fifth Avenue, Pittsburgh, PA 15213 USA ,0000 0004 0420 3665grid.413935.9VA Pittsburgh Healthcare System, Pittsburgh, PA USA
| | - Yohannes Tesfaigzi
- Lovelace Respiratory Research Institute, COPD program, Albuquerque, NM USA
| | - Tyrone Ryba
- 0000 0004 0504 9575grid.422569.eDivision of Natural Sciences, New College of Florida, Sarasota, FL USA
| | - Toru Nyunoya
- 0000 0004 1936 9000grid.21925.3dDepartment of Medicine, University of Pittsburgh, NW628 UPMC Montefiore, 3459 Fifth Avenue, Pittsburgh, PA 15213 USA ,0000 0004 0420 3665grid.413935.9VA Pittsburgh Healthcare System, Pittsburgh, PA USA
| |
Collapse
|
25
|
Xu Z, Che T, Li F, Tian K, Zhu Q, Mishra SK, Dai Y, Li M, Li D. The temporal expression patterns of brain transcriptome during chicken development and ageing. BMC Genomics 2018; 19:917. [PMID: 30545297 PMCID: PMC6293534 DOI: 10.1186/s12864-018-5301-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 11/21/2018] [Indexed: 12/12/2022] Open
Abstract
Background The transcriptional profiles of mammals during brain development and ageing have been characterized. However the global expression patterns of transcriptome in the chicken brain have not been explored. Here, we systematically investigated the temporal expression profiles of lncRNAs and mRNAs across 8 stages (including 3 embryonic stages, 2 growth stages and 3 adult stages) in the female chicken cerebrum. Results We identified 39,907 putative lncRNAs and 14,558 mRNAs, investigated the temporal expression patterns by tracking a set of age-dependent genes and predicted potential biological functions of lncRNAs based on co-expression network. The results showed that genes with functions in development, synapses and axons exhibited a progressive decay; genes related to immune response were up-regulated with age. Conclusions These results may reflect changes in the regulation of transcriptional networks and provide non-coding RNA gene candidates for further studies and would contribute to a comprehensive understanding of the molecular mechanisms of chicken development and may provide insights or deeper understanding regarding the regulatory mechanisms of age-dependent protein coding and non-protein coding genes in chicken. In addition, as the chicken is an important model organism bridging the evolutionary gap between mammals and other vertebrates, these high resolution data may provide a novel evidence to improve our comprehensive understanding of the brain transcriptome during vertebrate evolution. Electronic supplementary material The online version of this article (10.1186/s12864-018-5301-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhongxian Xu
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Tiandong Che
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Feng Li
- Key Laboratory of Southwest China Wildlife Resources Conservation (Ministry of Education), China West Normal University, Nanchong, 637009, Sichuan, China
| | - Kai Tian
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Qing Zhu
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Shailendra Kumar Mishra
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Yifei Dai
- Novogene Bioinformatics Institute, Beijing, 100083, China
| | - Mingzhou Li
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China.
| | - Diyan Li
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China.
| |
Collapse
|
26
|
Pennings JLA, Mariman R, Hodemaekers HM, Reemers SSN, Janssen R, Guichelaar T. Transcriptomics in lung tissue upon respiratory syncytial virus infection reveals aging as important modulator of immune activation and matrix maintenance. Sci Rep 2018; 8:16653. [PMID: 30413794 PMCID: PMC6226529 DOI: 10.1038/s41598-018-35180-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 10/30/2018] [Indexed: 12/25/2022] Open
Abstract
Aging poses an increased risk of severe infection by respiratory syncytial virus (RSV). The many different biological pathways comprising the response to infection in lungs that are influenced by aging are complex and remain to be defined more thoroughly. Towards finding new directions in research on aging, we aimed to define biological pathways in the acute response to RSV that are affected in the lungs by aging. We therefore profiled the full transcriptome of lung tissue of mice prior to and during RSV infection both at young and old age. In the absence of RSV, we found aging to downregulate genes that are involved in constitution of the extracellular matrix. Moreover, uninfected old mice showed elevated expression of pathways that resemble injury, metabolic aberrations, and disorders mediated by functions of the immune system that were induced at young age only by an exogenous trigger like RSV. Furthermore, infection by RSV mounted stronger activation of anti-viral type-I interferon pathways at old age. Despite such exaggerated anti-viral responses, old mice showed reduced control of virus. Altogether, our findings emphasize important roles in aging-related susceptibility to respiratory disease for extracellular matrix dysfunctions and dysregulated immune activation in lungs.
Collapse
Affiliation(s)
- Jeroen L A Pennings
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Rob Mariman
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Hennie M Hodemaekers
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Sylvia S N Reemers
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands.,MSD Animal Health, Boxmeer, The Netherlands
| | - Riny Janssen
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Teun Guichelaar
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands.
| |
Collapse
|
27
|
Mis MA, Rogers MF, Jeffries AR, Wilbrey AL, Chen L, Yang Y, Dib-Hajj S, Waxman SG, Stevens EB, Randall AD. Differential aging-related changes in neurophysiology and gene expression in IB4-positive and IB4-negative nociceptive neurons. Aging Cell 2018; 17:e12795. [PMID: 29943484 PMCID: PMC6052481 DOI: 10.1111/acel.12795] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 04/25/2018] [Accepted: 05/28/2018] [Indexed: 12/16/2022] Open
Abstract
Despite pain prevalence altering with age, the effects of aging on the properties of nociceptors are not well understood. Nociceptors, whose somas are located in dorsal root ganglia, are frequently divided into two groups based on their ability to bind isolectin B4 (IB4). Here, using cultured neurons from 1‐, 3‐, 5‐, 8‐, 12‐, and 18‐month‐old mice, we investigate age‐dependent changes in IB4‐positive and IB4‐negative neurons. Current‐clamp experiments at physiological temperature revealed nonlinear changes in firing frequency of IB4‐positive, but not IB4‐negative neurons, with a peak at 8 months. This was likely due to the presence of proexcitatory conductances activated at depolarized membrane potentials and significantly higher input resistances found in IB4‐positive neurons from 8‐month‐old mice. Repetitive firing in nociceptors is driven primarily by the TTX‐resistant sodium current, and indeed, IB4‐positive neurons from 8‐month‐old mice were found to receive larger contributions from the TTX‐resistant window current around the resting membrane potential. To further address the mechanisms behind these differences, we performed RNA‐seq experiments on IB4‐positive and IB4‐negative neurons from 1‐, 8‐, and 18‐month‐old mice. We found a larger number of genes significantly affected by age within the IB4‐positive than IB4‐negative neurons from 8‐month‐old mice, including known determinants of nociceptor excitability. The above pronounced age‐dependent changes at the cellular and molecular levels in IB4‐positive neurons point to potential mechanisms behind the reported increase in pain sensitivity in middle‐aged rodents and humans, and highlight the possibility of targeting a particular group of neurons in the development of age‐tailored pain treatments.
Collapse
Affiliation(s)
- Malgorzata A. Mis
- School of Physiology, Pharmacology, and Neuroscience; University of Bristol; Bristol UK
| | - Mark F. Rogers
- Intelligent Systems Laboratory; University of Bristol; Bristol UK
| | - Aaron R. Jeffries
- University of Exeter Medical School; University of Exeter; Exeter UK
| | | | - Lubin Chen
- Department of Neurology and Center for Neuroscience and Regeneration Research; Yale University School of Medicine; New Haven Connecticut USA
- Rehabilitation Research Center; Veterans Administration Connecticut Healthcare System; West Haven Connecticut USA
| | - Yang Yang
- Department of Medicinal Chemistry and Molecular Pharmacology; Purdue University College of Pharmacy and Purdue Institute for Integrative Neuroscience; West Lafayette Indiana USA
| | - Sulayman Dib-Hajj
- Department of Neurology and Center for Neuroscience and Regeneration Research; Yale University School of Medicine; New Haven Connecticut USA
- Rehabilitation Research Center; Veterans Administration Connecticut Healthcare System; West Haven Connecticut USA
| | - Stephen G. Waxman
- Department of Neurology and Center for Neuroscience and Regeneration Research; Yale University School of Medicine; New Haven Connecticut USA
- Rehabilitation Research Center; Veterans Administration Connecticut Healthcare System; West Haven Connecticut USA
| | | | - Andrew D. Randall
- School of Physiology, Pharmacology, and Neuroscience; University of Bristol; Bristol UK
- Institute of Biomedical and Clinical Sciences; University of Exeter Medical School; Hatherly Laboratories; University of Exeter; Exeter UK
| |
Collapse
|
28
|
Guichelaar T, van Erp EA, Hoeboer J, Smits NAM, van Els CACM, Pieren DKJ, Luytjes W. Diversity of aging of the immune system classified in the cotton rat (Sigmodon hispidus) model of human infectious diseases. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 82:39-48. [PMID: 29305168 DOI: 10.1016/j.dci.2017.12.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 12/28/2017] [Accepted: 12/29/2017] [Indexed: 06/07/2023]
Abstract
Susceptibility and declined resistance to human pathogens like respiratory syncytial virus (RSV) at old age is well represented in the cotton rat (Sigmodon hispidus). Despite providing a preferred model of human infectious diseases, little is known about aging of its adaptive immune system. We aimed to define aging-related changes of the immune system of this species. Concomitantly, we asked whether the rate of immunological alterations may be stratified by physiological aberrations encountered during aging. With increasing age, cotton rats showed reduced frequencies of T cells, impaired induction of antibodies to RSV, higher incidence of aberrations of organs and signs of lipemia. Moreover, old animals expressed high biological heterogeneity, but the age-related reduction of T cell frequency was only observed in those specimens that displayed aberrant organs. Thus, cotton rats show age-related alterations of lymphocytes that can be classified by links with health status.
Collapse
Affiliation(s)
- Teun Guichelaar
- Centre for Infectious Disease Control (CIb), National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands.
| | - Elisabeth A van Erp
- Centre for Infectious Disease Control (CIb), National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands; Laboratory of Pediatric Infectious Diseases, Department of Pediatrics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jeroen Hoeboer
- Centre for Infectious Disease Control (CIb), National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Noortje A M Smits
- Centre for Infectious Disease Control (CIb), National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Cécile A C M van Els
- Centre for Infectious Disease Control (CIb), National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Daan K J Pieren
- Centre for Infectious Disease Control (CIb), National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Willem Luytjes
- Centre for Infectious Disease Control (CIb), National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| |
Collapse
|
29
|
Antoch MP, Wrobel M, Kuropatwinski KK, Gitlin I, Leonova KI, Toshkov I, Gleiberman AS, Hutson AD, Chernova OB, Gudkov AV. Physiological frailty index (PFI): quantitative in-life estimate of individual biological age in mice. Aging (Albany NY) 2017; 9:615-626. [PMID: 28325885 PMCID: PMC5391222 DOI: 10.18632/aging.101206] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 03/11/2017] [Indexed: 11/25/2022]
Abstract
The development of healthspan-extending pharmaceuticals requires quantitative estimation of age-related progressive physiological decline. In humans, individual health status can be quantitatively assessed by means of a frailty index (FI), a parameter which reflects the scale of accumulation of age-related deficits. However, adaptation of this methodology to animal models is a challenging task since it includes multiple subjective parameters. Here we report a development of a quantitative non-invasive procedure to estimate biological age of an individual animal by creating physiological frailty index (PFI). We demonstrated the dynamics of PFI increase during chronological aging of male and female NIH Swiss mice. We also demonstrated acceleration of growth of PFI in animals placed on a high fat diet, reflecting aging acceleration by obesity and provide a tool for its quantitative assessment. Additionally, we showed that PFI could reveal anti-aging effect of mTOR inhibitor rapatar (bioavailable formulation of rapamycin) prior to registration of its effects on longevity. PFI revealed substantial sex-related differences in normal chronological aging and in the efficacy of detrimental (high fat diet) or beneficial (rapatar) aging modulatory factors. Together, these data introduce PFI as a reliable, non-invasive, quantitative tool suitable for testing potential anti-aging pharmaceuticals in pre-clinical studies.
Collapse
Affiliation(s)
- Marina P Antoch
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Michelle Wrobel
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA.,Everon Biosciences, Inc., Buffalo, NY 14203, USA
| | - Karen K Kuropatwinski
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Ilya Gitlin
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Katerina I Leonova
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Ilia Toshkov
- Everon Biosciences, Inc., Buffalo, NY 14203, USA
| | | | - Alan D Hutson
- Biostatistics and Bioinformatics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | | | - Andrei V Gudkov
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| |
Collapse
|
30
|
Li Z, Jiao Y, Fan EK, Scott MJ, Li Y, Li S, Billiar TR, Wilson MA, Shi X, Fan J. Aging-Impaired Filamentous Actin Polymerization Signaling Reduces Alveolar Macrophage Phagocytosis of Bacteria. THE JOURNAL OF IMMUNOLOGY 2017; 199:3176-3186. [PMID: 28947541 DOI: 10.4049/jimmunol.1700140] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 08/26/2017] [Indexed: 12/14/2022]
Abstract
In elderly patients, bacterial infection often causes severe complications and sepsis. Compared to younger patients, older patients are more susceptible to sepsis caused by respiratory infection. Macrophage (Mϕ) phagocytosis of bacteria plays a critical role in the clearance of pathogens and the initiation of immune responses. It has been suggested that Mϕ exhibit age-related functional alterations, including reduced chemotaxis, phagocytosis, antibacterial defense, and the ability to generate reactive oxygen species. However, the mechanisms behind these changes remain unclear. The present study sought to determine changes in bacterial phagocytosis in aging alveolar Mϕ (AMϕ) and the underlying mechanisms. We show that bacteria initiate cytoskeleton remodeling in AMϕ through interaction with macrophage receptor with collagenous structure (MARCO), a bacterial scavenger receptor. This remodeling, in turn, promotes enhanced cell surface expression of MARCO and bacterial phagocytosis. We further demonstrate that Rac1-GTP mediates MARCO signaling and activates actin-related protein-2/3 complex, an F-actin nucleator, thereby inducing F-actin polymerization, filopodia formation, and increased cell surface expression of MARCO, all of which are essential for the execution of bacteria phagocytosis. However, AMϕ isolated from aging mice exhibit suppressed Rac1 mRNA and protein expression, which resulted in decreases in Rac1-GTP levels and actin-related protein-2/3 activation, as well as subsequent attenuation of F-actin polymerization, filopodia formation, and cell surface expression of MARCO. As a result, bacterial phagocytosis in aging AMϕ is decreased. This study highlights a previously unidentified mechanism by which aging impairs Mϕ phagocytosis of bacteria. Targeting these pathways may improve outcomes of bacterial infection in elderly patients.
Collapse
Affiliation(s)
- Zhigang Li
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213.,Research and Development, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA 15240
| | - Yang Jiao
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213.,Department of Anesthesiology, Shanghai Xinhua Hospital, Jiaotong University School of Medicine, Shanghai 200092, China
| | - Erica K Fan
- University of Pittsburgh School of Arts and Science, Pittsburgh, PA 15213
| | - Melanie J Scott
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - Yuehua Li
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213.,Research and Development, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA 15240
| | - Song Li
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA 15261; and
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219
| | - Mark A Wilson
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213.,Research and Development, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA 15240
| | - Xueyin Shi
- Department of Anesthesiology, Shanghai Xinhua Hospital, Jiaotong University School of Medicine, Shanghai 200092, China;
| | - Jie Fan
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213; .,Research and Development, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA 15240.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219
| |
Collapse
|
31
|
Galbavy W, Lu Y, Kaczocha M, Puopolo M, Liu L, Rebecchi MJ. Transcriptomic evidence of a para-inflammatory state in the middle aged lumbar spinal cord. IMMUNITY & AGEING 2017; 14:9. [PMID: 28413428 PMCID: PMC5390443 DOI: 10.1186/s12979-017-0091-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 04/05/2017] [Indexed: 11/10/2022]
Abstract
BACKGROUND We have previously reported elevated expression of multiple pro-inflammatory markers in the lumbar spinal cord (LSC) of middle-aged male rats compared to young adults suggesting a para-inflammatory state develops in the LSC by middle age, a time that in humans is associated with the greatest pain prevalence and persistence. The goal of the current study was to examine the transcriptome-wide gene expression differences between young and middle aged LSC. METHODS Young (3 month) and middle-aged (17 month) naïve Fisher 344 rats (n = 5 per group) were euthanized, perfused with heparinized saline, and the LSC were removed. RESULTS ~70% of 31,000 coding sequences were detected. After normalization, ~ 1100 showed statistically significant differential expression. Of these genes, 353 middle-aged annotated genes differed by > 1.5 fold compared to the young group. Nearly 10% of these genes belonged to the microglial sensome. Analysis of this subset revealed that the principal age-related differential pathways populated are complement, pattern recognition receptors, OX40, and various T cell regulatory pathways consistent with microglial priming and T cell invasion and modulation. Many of these pathways substantially overlap those previously identified in studies of LSC of young animals with chronic inflammatory or neuropathic pain. CONCLUSIONS Up-modulation of complement pathway, microglial priming and activation, and T cell/antigen-presenting cell communication in healthy middle-aged LSC was found. Taken together with our previous work, the results support our conclusion that an incipient or para-inflammatory state develops in the LSC in healthy middle-aged adults.
Collapse
Affiliation(s)
- William Galbavy
- Department of Anesthesiology, School of Medicine, Health Sciences Center L4, Stony Brook University, Stony Brook, New York, 11794-8480 USA
| | - Yong Lu
- Department of Anesthesiology, School of Medicine, Health Sciences Center L4, Stony Brook University, Stony Brook, New York, 11794-8480 USA
| | - Martin Kaczocha
- Department of Anesthesiology, School of Medicine, Health Sciences Center L4, Stony Brook University, Stony Brook, New York, 11794-8480 USA
| | - Michelino Puopolo
- Department of Anesthesiology, School of Medicine, Health Sciences Center L4, Stony Brook University, Stony Brook, New York, 11794-8480 USA
| | - Lixin Liu
- Department of Anesthesiology, School of Medicine, Health Sciences Center L4, Stony Brook University, Stony Brook, New York, 11794-8480 USA
| | - Mario J Rebecchi
- Department of Anesthesiology, School of Medicine, Health Sciences Center L4, Stony Brook University, Stony Brook, New York, 11794-8480 USA
| |
Collapse
|
32
|
Braun F, Rinschen MM, Bartels V, Frommolt P, Habermann B, Hoeijmakers JHJ, Schumacher B, Dollé MET, Müller RU, Benzing T, Schermer B, Kurschat CE. Altered lipid metabolism in the aging kidney identified by three layered omic analysis. Aging (Albany NY) 2017; 8:441-57. [PMID: 26886165 PMCID: PMC4833139 DOI: 10.18632/aging.100900] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Aging-associated diseases and their comorbidities affect the life of a constantly growing proportion of the population in developed countries. At the center of these comorbidities are changes of kidney structure and function as age-related chronic kidney disease predisposes to the development of cardiovascular diseases such as stroke, myocardial infarction or heart failure. To detect molecular mechanisms involved in kidney aging, we analyzed gene expression profiles of kidneys from adult and aged wild-type mice by transcriptomic, proteomic and targeted lipidomic methodologies. Interestingly, transcriptome and proteome analyses revealed differential expression of genes primarily involved in lipid metabolism and immune response. Additional lipidomic analyses uncovered significant age-related differences in the total amount of phosphatidylethanolamines, phosphatidylcholines and sphingomyelins as well as in subspecies of phosphatidylserines and ceramides with age. By integration of these datasets we identified Aldh1a1, a key enzyme in vitamin A metabolism specifically expressed in the medullary ascending limb, as one of the most prominent upregulated proteins in old kidneys. Moreover, ceramidase Asah1 was highly expressed in aged kidneys, consistent with a decrease in ceramide C16. In summary, our data suggest that changes in lipid metabolism are involved in the process of kidney aging and in the development of chronic kidney disease.
Collapse
Affiliation(s)
- Fabian Braun
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Markus M Rinschen
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Valerie Bartels
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Department of Cardiology and Angiology, University of Münster, Münster, Germany
| | - Peter Frommolt
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Systems Biology of Ageing Cologne, University of Cologne, Cologne, Germany
| | - Bianca Habermann
- Systems Biology of Ageing Cologne, University of Cologne, Cologne, Germany.,Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Jan H J Hoeijmakers
- Department of Cell Biology and Genetics, Medical Genetics Centre, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Björn Schumacher
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Institute for Genome Stability in Aging and Disease, Medical Faculty, University of Cologne, Cologne, Germany
| | - Martijn E T Dollé
- National Institute of Public Health and the Environment, Centre for Health Protection, Bilthoven, The Netherlands
| | - Roman-Ulrich Müller
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Systems Biology of Ageing Cologne, University of Cologne, Cologne, Germany
| | - Thomas Benzing
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Systems Biology of Ageing Cologne, University of Cologne, Cologne, Germany
| | - Bernhard Schermer
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Systems Biology of Ageing Cologne, University of Cologne, Cologne, Germany
| | - Christine E Kurschat
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| |
Collapse
|
33
|
Investigating the specific core genetic-and-epigenetic networks of cellular mechanisms involved in human aging in peripheral blood mononuclear cells. Oncotarget 2017; 7:8556-79. [PMID: 26895224 PMCID: PMC4890987 DOI: 10.18632/oncotarget.7388] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 02/03/2016] [Indexed: 12/26/2022] Open
Abstract
Aging is an inevitable part of life for humans, and slowing down the aging process has become a main focus of human endeavor. Here, we applied a systems biology approach to construct protein-protein interaction networks, gene regulatory networks, and epigenetic networks, i.e. genetic and epigenetic networks (GENs), of elderly individuals and young controls. We then compared these GENs to extract aging mechanisms using microarray data in peripheral blood mononuclear cells, microRNA (miRNA) data, and database mining. The core GENs of elderly individuals and young controls were obtained by applying principal network projection to GENs based on Principal Component Analysis. By comparing the core networks, we identified that to overcome the accumulated mutation of genes in the aging process the transcription factor JUN can be activated by stress signals, including the MAPK signaling, T-cell receptor signaling, and neurotrophin signaling pathways through DNA methylation of BTG3, G0S2, and AP2B1 and the regulations of mir-223 let-7d, and mir-130a. We also address the aging mechanisms in old men and women. Furthermore, we proposed that drugs designed to target these DNA methylated genes or miRNAs may delay aging. A multiple drug combination comprising phenylalanine, cholesterol, and palbociclib was finally designed for delaying the aging process.
Collapse
|
34
|
Vermeij WP, Dollé MET, Reiling E, Jaarsma D, Payan-Gomez C, Bombardieri CR, Wu H, Roks AJM, Botter SM, van der Eerden BC, Youssef SA, Kuiper RV, Nagarajah B, van Oostrom CT, Brandt RMC, Barnhoorn S, Imholz S, Pennings JLA, de Bruin A, Gyenis Á, Pothof J, Vijg J, van Steeg H, Hoeijmakers JHJ. Restricted diet delays accelerated ageing and genomic stress in DNA-repair-deficient mice. Nature 2016; 537:427-431. [PMID: 27556946 PMCID: PMC5161687 DOI: 10.1038/nature19329] [Citation(s) in RCA: 213] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 07/25/2016] [Indexed: 12/27/2022]
Abstract
Mice deficient in the DNA excision-repair gene Ercc1 (Ercc1∆/-) show numerous accelerated ageing features that limit their lifespan to 4-6 months. They also exhibit a 'survival response', which suppresses growth and enhances cellular maintenance. Such a response resembles the anti-ageing response induced by dietary restriction (also known as caloric restriction). Here we report that a dietary restriction of 30% tripled the median and maximal remaining lifespans of these progeroid mice, strongly retarding numerous aspects of accelerated ageing. Mice undergoing dietary restriction retained 50% more neurons and maintained full motor function far beyond the lifespan of mice fed ad libitum. Other DNA-repair-deficient, progeroid Xpg-/- (also known as Ercc5-/-) mice, a model of Cockayne syndrome, responded similarly. The dietary restriction response in Ercc1∆/- mice closely resembled the effects of dietary restriction in wild-type animals. Notably, liver tissue from Ercc1∆/- mice fed ad libitum showed preferential extinction of the expression of long genes, a phenomenon we also observed in several tissues ageing normally. This is consistent with the accumulation of stochastic, transcription-blocking lesions that affect long genes more than short ones. Dietary restriction largely prevented this declining transcriptional output and reduced the number of γH2AX DNA damage foci, indicating that dietary restriction preserves genome function by alleviating DNA damage. Our findings establish the Ercc1∆/- mouse as a powerful model organism for health-sustaining interventions, reveal potential for reducing endogenous DNA damage, facilitate a better understanding of the molecular mechanism of dietary restriction and suggest a role for counterintuitive dietary-restriction-like therapy for human progeroid genome instability syndromes and possibly neurodegeneration in general.
Collapse
Affiliation(s)
- W P Vermeij
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - M E T Dollé
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), PO Box 1, 3720 BA Bilthoven, The Netherlands
| | - E Reiling
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands.,Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), PO Box 1, 3720 BA Bilthoven, The Netherlands
| | - D Jaarsma
- Department of Neuroscience, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - C Payan-Gomez
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands.,Facultad de Ciencias Naturales y Matemáticas, Universidad del Rosario, Carrera 24, 63C-69 Bogotá, Colombia
| | - C R Bombardieri
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - H Wu
- Department of Internal Medicine, Division of Vascular Medicine and Pharmacology, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - A J M Roks
- Department of Internal Medicine, Division of Vascular Medicine and Pharmacology, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - S M Botter
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands.,Laboratory for Orthopedic Research, Balgrist University Hospital, Forchstrasse 340, 8008, Zürich, Switzerland
| | - B C van der Eerden
- Department of Internal Medicine, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - S A Youssef
- Dutch Molecular Pathology Center, Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, PO Box 80125, 3508 TC Utrecht, The Netherlands
| | - R V Kuiper
- Dutch Molecular Pathology Center, Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, PO Box 80125, 3508 TC Utrecht, The Netherlands
| | - B Nagarajah
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), PO Box 1, 3720 BA Bilthoven, The Netherlands
| | - C T van Oostrom
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), PO Box 1, 3720 BA Bilthoven, The Netherlands
| | - R M C Brandt
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - S Barnhoorn
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - S Imholz
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), PO Box 1, 3720 BA Bilthoven, The Netherlands
| | - J L A Pennings
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), PO Box 1, 3720 BA Bilthoven, The Netherlands
| | - A de Bruin
- Dutch Molecular Pathology Center, Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, PO Box 80125, 3508 TC Utrecht, The Netherlands.,Department of Pediatrics, Division Molecular Genetics, University Medical Center Groningen, PO Box 30001, 9700 RB Groningen, The Netherlands
| | - Á Gyenis
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - J Pothof
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - J Vijg
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York 10461, USA
| | - H van Steeg
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), PO Box 1, 3720 BA Bilthoven, The Netherlands.,Department of Human Genetics, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - J H J Hoeijmakers
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands.,CECAD Forschungszentrum, Universität zu Köln, Joseph-Stelzmann-Straße 26, 50931 Köln, Germany
| |
Collapse
|
35
|
Haustead DJ, Stevenson A, Saxena V, Marriage F, Firth M, Silla R, Martin L, Adcroft KF, Rea S, Day PJ, Melton P, Wood FM, Fear MW. Transcriptome analysis of human ageing in male skin shows mid-life period of variability and central role of NF-κB. Sci Rep 2016; 6:26846. [PMID: 27229172 PMCID: PMC4882522 DOI: 10.1038/srep26846] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 05/09/2016] [Indexed: 01/21/2023] Open
Abstract
Age is well-known to be a significant factor in both disease pathology and response to treatment, yet the molecular changes that occur with age in humans remain ill-defined. Here, using transcriptome profiling of healthy human male skin, we demonstrate that there is a period of significantly elevated, transcriptome-wide expression changes occurring predominantly in middle age. Both pre and post this period, the transcriptome appears to undergo much smaller, linear changes with increasing age. Functional analysis of the transient changes in middle age suggest a period of heightened metabolic activity and cellular damage associated with NF-kappa-B and TNF signaling pathways. Through meta-analysis we also show the presence of global, tissue independent linear transcriptome changes with age which appear to be regulated by NF-kappa-B. These results suggest that aging in human skin is associated with a critical mid-life period with widespread transcriptome changes, both preceded and proceeded by a relatively steady rate of linear change in the transcriptome. The data provides insight into molecular changes associated with normal aging and will help to better understand the increasingly important pathological changes associated with aging.
Collapse
Affiliation(s)
- Daniel J. Haustead
- The Fiona Wood Foundation, Perth, WA 6000, Australia
- Faculty of Medicine, Dentistry and Health Sciences, University of Western Australia, Crawley WA 6009, Australia
- Faculty of Medicine and Health Sciences, University of Manchester, Manchester, M1 7DN, UK
| | - Andrew Stevenson
- The Fiona Wood Foundation, Perth, WA 6000, Australia
- Burn Injury Research Unit, School of Surgery, University of Western Australia, Crawley WA 6009, Australia
| | - Vishal Saxena
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Fiona Marriage
- Faculty of Medicine and Health Sciences, University of Manchester, Manchester, M1 7DN, UK
| | - Martin Firth
- Faculty of Medicine, Dentistry and Health Sciences, University of Western Australia, Crawley WA 6009, Australia
| | - Robyn Silla
- The Fiona Wood Foundation, Perth, WA 6000, Australia
- Burns Service of Western Australia, Royal Perth Hospital and Princess Margaret Hospital, WA 6000, Australia
| | - Lisa Martin
- The Fiona Wood Foundation, Perth, WA 6000, Australia
- Burns Service of Western Australia, Royal Perth Hospital and Princess Margaret Hospital, WA 6000, Australia
| | - Katharine F. Adcroft
- The Fiona Wood Foundation, Perth, WA 6000, Australia
- Burn Injury Research Unit, School of Surgery, University of Western Australia, Crawley WA 6009, Australia
| | - Suzanne Rea
- The Fiona Wood Foundation, Perth, WA 6000, Australia
- Burn Injury Research Unit, School of Surgery, University of Western Australia, Crawley WA 6009, Australia
- Burns Service of Western Australia, Royal Perth Hospital and Princess Margaret Hospital, WA 6000, Australia
| | - Philip J. Day
- Faculty of Medicine and Health Sciences, University of Manchester, Manchester, M1 7DN, UK
| | - Phillip Melton
- Centre for Genetic Origins of Health and Disease, University of Western Australia, Crawley WA , Australia 6009
| | - Fiona M. Wood
- The Fiona Wood Foundation, Perth, WA 6000, Australia
- Faculty of Medicine, Dentistry and Health Sciences, University of Western Australia, Crawley WA 6009, Australia
- Burn Injury Research Unit, School of Surgery, University of Western Australia, Crawley WA 6009, Australia
- Burns Service of Western Australia, Royal Perth Hospital and Princess Margaret Hospital, WA 6000, Australia
| | - Mark W. Fear
- The Fiona Wood Foundation, Perth, WA 6000, Australia
- Burn Injury Research Unit, School of Surgery, University of Western Australia, Crawley WA 6009, Australia
| |
Collapse
|
36
|
Abstract
Aging is a risk factor for several of the world's most prevalent diseases, including neurodegenerative disorders, cancer, cardiovascular disease and metabolic disease. Although our understanding of the molecular pathways that contribute to the aging process and age-related disease is progressing through the use of model organisms, how to apply this knowledge in the clinic is less clear. In September, Nature Medicine, in collaboration with the Volkswagen Foundation, hosted a conference at the beautiful Herrenhausen Palace in Hannover, Germany with the goal of broadening our understanding of the aging process and its meaning as a 'risk factor' in disease. Here, several of the speakers at that conference answer questions posed by Nature Medicine.
Collapse
|
37
|
Abstract
Progeroid mouse models display phenotypes in multiple organ systems that suggest premature aging and resemble features of natural aging of both mice and humans. The prospect of a significant increase in the global elderly population within the next decades has led to the emergence of "geroscience," which aims at elucidating the molecular mechanisms involved in aging. Progeroid mouse models are frequently used in geroscience as they provide insight into the molecular mechanisms that are involved in the highly complex process of natural aging. This review provides an overview of the most commonly reported nonneoplastic macroscopic and microscopic pathologic findings in progeroid mouse models (eg, osteoporosis, osteoarthritis, degenerative joint disease, intervertebral disc degeneration, kyphosis, sarcopenia, cutaneous atrophy, wound healing, hair loss, alopecia, lymphoid atrophy, cataract, corneal endothelial dystrophy, retinal degenerative diseases, and vascular remodeling). Furthermore, several shortcomings in pathologic analysis and descriptions of these models are discussed. Progeroid mouse models are valuable models for aging, but thorough knowledge of both the mouse strain background and the progeria-related phenotype is required to guide interpretation and translation of the pathology data.
Collapse
Affiliation(s)
- L Harkema
- Dutch Molecular Pathology Center, Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - S A Youssef
- Dutch Molecular Pathology Center, Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - A de Bruin
- Dutch Molecular Pathology Center, Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands Department of Pediatrics, Division of Molecular Genetics, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
38
|
Confounding Factors in the Transcriptome Analysis of an In-Vivo Exposure Experiment. PLoS One 2016; 11:e0145252. [PMID: 26789003 PMCID: PMC4720430 DOI: 10.1371/journal.pone.0145252] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 11/30/2015] [Indexed: 11/19/2022] Open
Abstract
Confounding factors In transcriptomics experimentation, confounding factors frequently exist alongside the intended experimental factors and can severely influence the outcome of a transcriptome analysis. Confounding factors are regularly discussed in methodological literature, but their actual, practical impact on the outcome and interpretation of transcriptomics experiments is, to our knowledge, not documented. For instance, in-vivo experimental factors; like Individual, Sample-Composition and Time-of-Day are potentially formidable confounding factors. To study these confounding factors, we designed an extensive in-vivo transcriptome experiment (n = 264) with UVR exposure of murine skin containing six consecutive samples from each individual mouse (n = 64). Analysis Approach Evaluation of the confounding factors: Sample-Composition, Time-of-Day, Handling-Stress, and Individual-Mouse resulted in the identification of many genes that were affected by them. These genes sometimes showed over 30-fold expression differences. The most prominent confounding factor was Sample-Composition caused by mouse-dependent skin composition differences, sampling variation and/or influx/efflux of mobile cells. Although we can only evaluate these effects for known cell type specifically expressed genes in our complex heterogeneous samples, it is clear that the observed variations also affect the cumulative expression levels of many other non-cell-type-specific genes. ANOVA ANOVA analysis can only attempt to neutralize the effects of the well-defined confounding factors, such as Individual-Mouse, on the experimental factors UV-Dose and Recovery-Time. Also, by definition, ANOVA only yields reproducible gene-expression differences, but we found that these differences were very small compared to the fold changes induced by the confounding factors, questioning the biological relevance of these ANOVA-detected differences. Furthermore, it turned out that many of the differentially expressed genes found by ANOVA were also present in the gene clusters associated with the confounding factors. Conclusion Hence our overall conclusion is that confounding factors have a major impact on the outcome of in-vivo transcriptomics experiments. Thus the set-up, analysis, and interpretation of such experiments should be approached with the utmost prudence.
Collapse
|
39
|
Rangaraju S, Solis GM, Thompson RC, Gomez-Amaro RL, Kurian L, Encalada SE, Niculescu AB, Salomon DR, Petrascheck M. Suppression of transcriptional drift extends C. elegans lifespan by postponing the onset of mortality. eLife 2015; 4:e08833. [PMID: 26623667 PMCID: PMC4720515 DOI: 10.7554/elife.08833] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 10/29/2015] [Indexed: 12/28/2022] Open
Abstract
Longevity mechanisms increase lifespan by counteracting the effects of aging. However, whether longevity mechanisms counteract the effects of aging continually throughout life, or whether they act during specific periods of life, preventing changes that precede mortality is unclear. Here, we uncover transcriptional drift, a phenomenon that describes how aging causes genes within functional groups to change expression in opposing directions. These changes cause a transcriptome-wide loss in mRNA stoichiometry and loss of co-expression patterns in aging animals, as compared to young adults. Using Caenorhabditis elegans as a model, we show that extending lifespan by inhibiting serotonergic signals by the antidepressant mianserin attenuates transcriptional drift, allowing the preservation of a younger transcriptome into an older age. Our data are consistent with a model in which inhibition of serotonergic signals slows age-dependent physiological decline and the associated rise in mortality levels exclusively in young adults, thereby postponing the onset of major mortality. DOI:http://dx.doi.org/10.7554/eLife.08833.001 All organisms age, leading to gradual declines in the body’s systems and eventually death. How certain genetic mutations and drugs delay the effects of aging and promote survival to an older age is a question many researchers are exploring. One way this problem is investigated is by looking at how the activity – or expression – of different genes changes during aging. Scientists interested in understanding aging and longevity often study a simple worm called Caenorhabditis elegans. This worm normally lives for about three weeks, and young C. elegans are able to produce offspring within days of hatching. This accelerated life cycle allows scientists to observe the entire lifespan of the worms. Over time, experiments have shown that DNA damage, changes in behavior and changes to gene expression are all markers of aging in the worms. Now, Rangaraju et al. describe how changes in gene expression patterns that begin early in the lives of C. elegans shorten their lifespan. Specifically, in groups of genes that work together, some genes increase expression, while others decrease expression with age. This phenomenon is called “transcriptional drift” and leads to an age-associated loss of coordination among groups of genes that help orchestrate specific tasks. Rangaraju et al. show that an antidepressant called mianserin prevents transcriptional drift in many of C. elegans’ genes: young worms treated with the drug resist the effects of aging on the transcriptome and maintain coordinated patterns of gene expression for longer. Maintaining coordinated patterns of gene expression postpones the onset of age-related bodily declines and extends the life of treated worms by extending the duration of young adulthood and postponing the onset of age-associated death. The drug also appears to protect against stress-induced changes in gene expression. This suggests that some of the age-related shifts in gene expression occur when cells fail to recover normal gene expression patterns after a stressful event. Questions that remain to be investigated in future studies are whether other longevity mechanisms also extend lifespan by preserving coordinated gene expression patterns, and whether other longevity mechanisms act by extending specific periods of life. DOI:http://dx.doi.org/10.7554/eLife.08833.002
Collapse
Affiliation(s)
- Sunitha Rangaraju
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, United States.,Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, United States.,Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, United States.,Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, United States
| | - Gregory M Solis
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, United States.,Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, United States.,Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, United States.,Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, United States
| | - Ryan C Thompson
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, United States
| | - Rafael L Gomez-Amaro
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, United States.,Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, United States.,Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, United States.,Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, United States
| | - Leo Kurian
- Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | - Sandra E Encalada
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, United States.,Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, United States.,Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, United States
| | - Alexander B Niculescu
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, United States
| | - Daniel R Salomon
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, United States
| | - Michael Petrascheck
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, United States.,Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, United States.,Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, United States.,Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, United States
| |
Collapse
|
40
|
White RR, Milholland B, MacRae SL, Lin M, Zheng D, Vijg J. Comprehensive transcriptional landscape of aging mouse liver. BMC Genomics 2015; 16:899. [PMID: 26541291 PMCID: PMC4636074 DOI: 10.1186/s12864-015-2061-8] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 10/10/2015] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Mammalian aging is a highly complex process, a full mechanistic understanding of which is still lacking. One way to help understand the molecular changes underlying aging is through a comprehensive analysis of the transcriptome, the primary determinant of age-related phenotypic diversity. Previous studies have relied on microarray analysis to examine gene expression profiles in different tissues of aging organisms. However, studies have shown microarray-based transcriptional profiling is less accurate and not fully capable of capturing certain intricacies of the global transcriptome. METHODS Here, using directional whole transcriptome RNA-sequencing of aged mouse liver we have identified a comprehensive high-resolution profile of differentially expressed liver transcripts comprised of canonical protein-coding transcripts, transcript isoforms, and non-coding RNA transcripts, including pseudogenes, long non-coding RNAs and small RNA species. RESULTS Results show extensive age-related changes in every component of the mouse liver transcriptome and a pronounced increase in inter-individual variation. Functional annotation of the protein-coding mRNAs and isoforms indicated broad alterations in immune response, cell activation, metabolic processes, and RNA modification. Interestingly, multiple lncRNAs (Meg3, Rian, Mirg) from the Dlk-Dio3 microRNA locus were found up-regulated in aging liver, classifying this locus as a putative regulatory hotspot locus in aging liver. Moreover, integration of the altered non-coding RNAs and protein-coding transcripts into interaction networks of age-related change revealed inflammation, cellular proliferation, and metabolism as the dominant aging phenotypes in mouse liver. CONCLUSIONS Our analyses provide the first comprehensive dissection of the transcriptional landscape in aging mouse liver.
Collapse
Affiliation(s)
- Ryan R White
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA. .,Albert Einstein College of Medicine, Michael F. Price Center for Genetic and Translational Research, 1301 Morris Park Ave, Bronx, NY, 10461, USA.
| | - Brandon Milholland
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Sheila L MacRae
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Mingyan Lin
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Deyou Zheng
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA. .,Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Jan Vijg
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA. .,Albert Einstein College of Medicine, Michael F. Price Center for Genetic and Translational Research, 1301 Morris Park Ave, Bronx, NY, 10461, USA.
| |
Collapse
|
41
|
Abstract
Several studies show evidence for the genetic basis of renal disease, which renders some individuals more prone than others to accelerated renal aging. Studying the genetics of renal aging can help us to identify genes involved in this process and to unravel the underlying pathways. First, this opinion article will give an overview of the phenotypes that can be observed in age-related kidney disease. Accurate phenotyping is essential in performing genetic analysis. For kidney aging, this could include both functional and structural changes. Subsequently, this article reviews the studies that report on candidate genes associated with renal aging in humans and mice. Several loci or candidate genes have been found associated with kidney disease, but identification of the specific genetic variants involved has proven to be difficult. CUBN, UMOD, and SHROOM3 were identified by human GWAS as being associated with albuminuria, kidney function, and chronic kidney disease (CKD). These are promising examples of genes that could be involved in renal aging, and were further mechanistically evaluated in animal models. Eventually, we will provide approaches for performing genetic analysis. We should leverage the power of mouse models, as testing in humans is limited. Mouse and other animal models can be used to explain the underlying biological mechanisms of genes and loci identified by human GWAS. Furthermore, mouse models can be used to identify genetic variants associated with age-associated histological changes, of which Far2, Wisp2, and Esrrg are examples. A new outbred mouse population with high genetic diversity will facilitate the identification of genes associated with renal aging by enabling high-resolution genetic mapping while also allowing the control of environmental factors, and by enabling access to renal tissues at specific time points for histology, proteomics, and gene expression.
Collapse
Affiliation(s)
- Gerda A. Noordmans
- Department of Pathology and Medical Biology University of Groningen University Medical Center Groningen Groningen the Netherlands
| | - Jan‐Luuk Hillebrands
- Department of Pathology and Medical Biology University of Groningen University Medical Center Groningen Groningen the Netherlands
| | - Harry Goor
- Department of Pathology and Medical Biology University of Groningen University Medical Center Groningen Groningen the Netherlands
| | | |
Collapse
|
42
|
Abstract
The 2014 joint meeting of the International Society for Cellular Oncology (ISCO) and the European Workshop on Cytogenetics and Molecular Genetics of Solid Tumors (EWCMST), organized by Nick Gilbert, Juan Cigudosa and Bauke Ylstra, was held from 11 to 14 May in Malaga, Spain. Since the previous meeting in 2012, the ever increasing availability of new sequencing technologies has enabled the analysis of cancer genomes at an increasingly greater detail. In addition to structural changes in the genome (i.e., translocations, deletions, amplifications), frequent mutations in important regulatory genes have been found to occur, as also frequent alterations in a large number of epigenetic factors. The challenge now is to relate structural changes in cancer genomes to the underlying disease mechanisms and to reveal opportunities for the design of novel (targeted) therapies. During the meeting, various topics related to these challenges and opportunities were addressed, including those dealing with functional genomics, genome instability, biomarkers and diagnostics, cancer genetics and epigenomics. Special attention was paid to therapy-driven cancer evolution (keynote lecture) and relationships between DNA repair, cancer and ageing (Prof. Ploem lecture). Based on the information presented at the meeting, several aspects of the cancer genome and its functional implications are provided in this report.
Collapse
|
43
|
Controlled induction of DNA double-strand breaks in the mouse liver induces features of tissue ageing. Nat Commun 2015; 6:6790. [PMID: 25858675 PMCID: PMC4394211 DOI: 10.1038/ncomms7790] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 02/27/2015] [Indexed: 01/07/2023] Open
Abstract
DNA damage has been implicated in ageing, but direct evidence for a causal relationship is lacking, owing to the difficulty of inducing defined DNA lesions in cells and tissues without simultaneously damaging other biomolecules and cellular structures. Here we directly test whether highly toxic DNA double-strand breaks (DSBs) alone can drive an ageing phenotype using an adenovirus-based system based on tetracycline-controlled expression of the SacI restriction enzyme. We deliver the adenovirus to mice and compare molecular and cellular end points in the liver with normally aged animals. Treated, 3-month-old mice display many, but not all signs of normal liver ageing as early as 1 month after treatment, including ageing pathologies, markers of senescence, fused mitochondria and alterations in gene expression profiles. These results, showing that DSBs alone can cause distinct ageing phenotypes in mouse liver, provide new insights in the role of DNA damage as a driver of tissue ageing. Accumulation of DNA damage is a hallmark of cellular ageing but cause and effect are unclear. Here White et al. induce clean DNA double-strand breaks in the liver of mice using a modified restriction enzyme and demonstrate that DNA damage alone is sufficient to recapitulate some aspects of tissue ageing.
Collapse
|
44
|
Ng'oma E, Reichwald K, Dorn A, Wittig M, Balschun T, Franke A, Platzer M, Cellerino A. The age related markers lipofuscin and apoptosis show different genetic architecture by QTL mapping in short-lived Nothobranchius fish. Aging (Albany NY) 2015; 6:468-80. [PMID: 25093339 PMCID: PMC4100809 DOI: 10.18632/aging.100660] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Annual fish of the genus Nothobranchius show large variations in lifespan and expression of age-related phenotypes between closely related populations. We studied N. kadleci and its sister species N. furzeri GRZ strain, and found that N.kadleci is longer-lived than the N. furzeri. Lipofuscin and apoptosis measured in the liver increased with age in N. kadleci with different profiles: lipofuscin increased linearly, while apoptosis declined in the oldest animals. More lipofuscin (P<0.001) and apoptosis (P<0.001) was observed in N. furzeri than in N. kadleci at 16w age. Lipofuscin and apoptotic cells were then quantified in hybrids from the mating of N. furzeri to N. kadleci. F₁individuals showed heterosis for lipofuscin but additive effects for apoptosis. These two age-related phenotypes were not correlated in F₂ hybrids. Quantitative trait loci analysis of 287 F₂ fish using 237 markers identified two QTL accounting for 10% of lipofuscin variance (P<0.001) with overdominance effect. Apoptotic cells revealed three significant- and two suggestive QTL explaining 19% of variance (P<0.001), showing additive and dominance effects, and two interacting loci. Our results show that lipofuscin and apoptosis are markers of different age-dependent biological processes controlled by different genetic mechanisms.
Collapse
Affiliation(s)
- Enoch Ng'oma
- Biology of Ageing, Leibniz Institute for Age Research - Fritz Lipmann Institute, 07745 Jena, Germany
| | - Kathrin Reichwald
- Genome Analysis, Leibniz Institute for Age Research - Fritz Lipmann Institute, 07745 Jena, Germany
| | - Alexander Dorn
- Biology of Ageing, Leibniz Institute for Age Research - Fritz Lipmann Institute, 07745 Jena, Germany
| | - Michael Wittig
- Institute of Clinical Molecular Biology, Christian-Albrechts-University, 24105 Kiel, Germany
| | - Tobias Balschun
- Hufeland Klinikum Mühlhausen, Institut für Infektiologie und Pathobiologie, 99974 Mühlhausen, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University, 24105 Kiel, Germany
| | - Matthias Platzer
- Genome Analysis, Leibniz Institute for Age Research - Fritz Lipmann Institute, 07745 Jena, Germany
| | - Allesandro Cellerino
- Biology of Ageing, Leibniz Institute for Age Research - Fritz Lipmann Institute, 07745 Jena, Germany. Neurobiology Laboratory, Scuola Normale Superiore, 56124 Pisa, Italy
| |
Collapse
|
45
|
Baumgart M, Groth M, Priebe S, Savino A, Testa G, Dix A, Ripa R, Spallotta F, Gaetano C, Ori M, Terzibasi Tozzini E, Guthke R, Platzer M, Cellerino A. RNA-seq of the aging brain in the short-lived fish N. furzeri - conserved pathways and novel genes associated with neurogenesis. Aging Cell 2014; 13:965-74. [PMID: 25059688 PMCID: PMC4326923 DOI: 10.1111/acel.12257] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2014] [Indexed: 11/30/2022] Open
Abstract
The brains of teleost fish show extensive adult neurogenesis and neuronal regeneration. The patterns of gene regulation during fish brain aging are unknown. The short-lived teleost fish Nothobranchius furzeri shows markers of brain aging including reduced learning performances, gliosis, and reduced adult neurogenesis. We used RNA-seq to quantify genome-wide transcript regulation and sampled five different time points to characterize whole-genome transcript regulation during brain aging of N. furzeri. Comparison with human datasets revealed conserved up-regulation of ribosome, lysosome, and complement activation and conserved down-regulation of synapse, mitochondrion, proteasome, and spliceosome. Down-regulated genes differ in their temporal profiles: neurogenesis and extracellular matrix genes showed rapid decay, synaptic and axonal genes a progressive decay. A substantial proportion of differentially expressed genes (∼40%) showed inversion of their temporal profiles in the last time point: spliceosome and proteasome showed initial down-regulation and stress-response genes initial up-regulation. Extensive regulation was detected for chromatin remodelers of the DNMT and CBX families as well as members of the polycomb complex and was mirrored by an up-regulation of the H3K27me3 epigenetic mark. Network analysis showed extensive coregulation of cell cycle/DNA synthesis genes with the uncharacterized zinc-finger protein ZNF367 as central hub. In situ hybridization showed that ZNF367 is expressed in neuronal stem cell niches of both embryonic zebrafish and adult N. furzeri. Other genes down-regulated with age, not previously associated with adult neurogenesis and with similar patterns of expression are AGR2, DNMT3A, KRCP, MEX3A, SCML4, and CBX1. CBX7, on the other hand, was up-regulated with age.
Collapse
Affiliation(s)
- Mario Baumgart
- Leibniz Institute for Age Research - Fritz Lipmann Institute e.V. (FLI); Jena Germany
| | - Marco Groth
- Leibniz Institute for Age Research - Fritz Lipmann Institute e.V. (FLI); Jena Germany
| | - Steffen Priebe
- Leibniz Institute for Natural Product Research and Infection Biology - Hans-Knöll-Institute e.V. (HKI); Jena Germany
| | - Aurora Savino
- Laboratory of NeuroBiology; Scuola Normale Superiore; University of Pisa; Pisa Italy
| | - Giovanna Testa
- Laboratory of NeuroBiology; Scuola Normale Superiore; University of Pisa; Pisa Italy
- Department of Biology; University of Pisa; Pisa Italy
| | - Andreas Dix
- Leibniz Institute for Natural Product Research and Infection Biology - Hans-Knöll-Institute e.V. (HKI); Jena Germany
| | - Roberto Ripa
- Laboratory of NeuroBiology; Scuola Normale Superiore; University of Pisa; Pisa Italy
| | | | - Carlo Gaetano
- Klinikum der Johann Wolfgang Goethe-Universität; Frankfurt am Main Germany
| | - Michela Ori
- Department of Biology; University of Pisa; Pisa Italy
| | - Eva Terzibasi Tozzini
- Laboratory of NeuroBiology; Scuola Normale Superiore; University of Pisa; Pisa Italy
| | - Reinhard Guthke
- Leibniz Institute for Natural Product Research and Infection Biology - Hans-Knöll-Institute e.V. (HKI); Jena Germany
| | - Matthias Platzer
- Leibniz Institute for Age Research - Fritz Lipmann Institute e.V. (FLI); Jena Germany
| | - Alessandro Cellerino
- Leibniz Institute for Age Research - Fritz Lipmann Institute e.V. (FLI); Jena Germany
- Laboratory of NeuroBiology; Scuola Normale Superiore; University of Pisa; Pisa Italy
| |
Collapse
|
46
|
Melis JPM, Jonker MJ, Vijg J, Hoeijmakers JHJ, Breit TM, van Steeg H. Aging on a different scale--chronological versus pathology-related aging. Aging (Albany NY) 2014; 5:782-8. [PMID: 24131799 PMCID: PMC3838780 DOI: 10.18632/aging.100606] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In the next decades the elderly population will increase dramatically, demanding appropriate solutions in health care and aging research focusing on healthy aging to prevent high burdens and costs in health care. For this, research targeting tissue-specific and individual aging is paramount to make the necessary progression in aging research. In a recently published study we have attempted to make a step interpreting aging data on chronological as well as pathological scale. For this, we sampled five major tissues at regular time intervals during the entire C57BL/6J murine lifespan from a controlled in vivo aging study, measured the whole transcriptome and incorporated temporal as well as physical health aspects into the analyses. In total, we used 18 different age-related pathological parameters and transcriptomic profiles of liver, kidney, spleen, lung and brain and created a database that can now be used for a broad systems biology approach. In our study, we focused on the dynamics of biological processes during chronological aging and the comparison between chronological and pathology-related aging.
Collapse
Affiliation(s)
- Joost P M Melis
- National Institute for Public Health and the Environment (RIVM), Center for Health Protection, Bilthoven, the Netherlands
| | | | | | | | | | | |
Collapse
|
47
|
Avanesov AS, Ma S, Pierce KA, Yim SH, Lee BC, Clish CB, Gladyshev VN. Age- and diet-associated metabolome remodeling characterizes the aging process driven by damage accumulation. eLife 2014; 3:e02077. [PMID: 24843015 PMCID: PMC4003482 DOI: 10.7554/elife.02077] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Aging is thought to be associated with increased molecular damage, but representative markers vary across conditions and organisms, making it difficult to assess properties of cumulative damage throughout lifespan. We used nontargeted metabolite profiling to follow age-associated trajectories of >15,000 metabolites in Drosophila subjected to control and lifespan-extending diets. We find that aging is associated with increased metabolite diversity and low-abundance molecules, suggesting they include cumulative damage. Remarkably, the number of detected compounds leveled-off in late-life, and this pattern associated with survivorship. Fourteen percent of metabolites showed age-associated changes, which decelerated in late-life and long-lived flies. In contrast, known metabolites changed in abundance similarly to nontargeted metabolites and transcripts, but did not increase in diversity. Targeted profiling also revealed slower metabolism and accumulation of lifespan-limiting molecules. Thus, aging is characterized by gradual metabolome remodeling, and condition- and advanced age-associated deceleration of this remodeling is linked to mortality and molecular damage.DOI: http://dx.doi.org/10.7554/eLife.02077.001.
Collapse
Affiliation(s)
- Andrei S Avanesov
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | - Siming Ma
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | | | - Sun Hee Yim
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | - Byung Cheon Lee
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | | | - Vadim N Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, United States Broad Institute, Cambridge, United States
| |
Collapse
|
48
|
Melis JPM, Derks KWJ, Pronk TE, Wackers P, Schaap MM, Zwart E, van Ijcken WFJ, Jonker MJ, Breit TM, Pothof J, van Steeg H, Luijten M. In vivo murine hepatic microRNA and mRNA expression signatures predicting the (non-)genotoxic carcinogenic potential of chemicals. Arch Toxicol 2014; 88:1023-34. [PMID: 24390151 DOI: 10.1007/s00204-013-1189-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 12/18/2013] [Indexed: 01/06/2023]
Abstract
There is a high need to improve the assessment of, especially non-genotoxic, carcinogenic features of chemicals. We therefore explored a toxicogenomics-based approach using genome-wide microRNA and mRNA expression profiles upon short-term exposure in mice. For this, wild-type mice were exposed for seven days to three different classes of chemicals, i.e., four genotoxic carcinogens (GTXC), seven non-genotoxic carcinogens (NGTXC), and five toxic non-carcinogens. Hepatic expression patterns of mRNA and microRNA transcripts were determined after exposure and used to assess the discriminative power of the in vivo transcriptome for GTXC and NGTXC. A final classifier set, discriminative for GTXC and NGTXC, was generated from the transcriptomic data using a tiered approach. This appeared to be a valid approach, since the predictive power of the final classifier set in three different classifier algorithms was very high for the original training set of chemicals. Subsequent validation in an additional set of chemicals revealed that the predictive power for GTXC remained high, in contrast to NGTXC, which appeared to be more troublesome. Our study demonstrated that the in vivo microRNA-ome has less discriminative power to correctly identify (non-)genotoxic carcinogen classes. The results generally indicate that single mRNA transcripts do have the potential to be applied in risk assessment, but that additional (genomic) strategies are necessary to correctly predict the non-genotoxic carcinogenic potential of a chemical.
Collapse
Affiliation(s)
- Joost P M Melis
- Center for Health Protection, National Institute for Public Health and the Environment (RIVM), P.O. Box 1, 3720 BA, Bilthoven, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|